Skip to main content
Diabetes Care logoLink to Diabetes Care
. 2022 Dec 12;46(Suppl 1):S158–S190. doi: 10.2337/dc23-S010

10. Cardiovascular Disease and Risk Management: Standards of Care in Diabetes—2023

Nuha A ElSayed, Grazia Aleppo, Vanita R Aroda, Raveendhara R Bannuru, Florence M Brown, Dennis Bruemmer, Billy S Collins, Sandeep R Das, Marisa E Hilliard, Diana Isaacs, Eric L Johnson, Scott Kahan, Kamlesh Khunti, Mikhail Kosiborod, Jose Leon, Sarah K Lyons, Mary Lou Perry, Priya Prahalad, Richard E Pratley, Jane Jeffrie Seley, Robert C Stanton, Robert A Gabbay, on behalf of the American Diabetes Association
PMCID: PMC9810475  PMID: 36507632

Abstract

The American Diabetes Association (ADA) “Standards of 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, 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 and a full list of Professional Practice Committee members, please refer to Introduction and Methodology. Readers who wish to comment on the Standards of Care are invited to do so at professional.diabetes.org/SOC.


For prevention and management of diabetes complications in children and adolescents, please refer to Section 14, “Children and Adolescents.”

Atherosclerotic cardiovascular disease (ASCVD)—defined as coronary heart disease (CHD), cerebrovascular disease, or peripheral arterial disease presumed to be of atherosclerotic origin—is the leading cause of morbidity and mortality for individuals with diabetes and results in an estimated $37.3 billion in cardiovascular-related spending per year associated with diabetes (1). Common conditions coexisting with type 2 diabetes (e.g., hypertension and dyslipidemia) are clear risk factors for ASCVD, and diabetes itself confers independent risk. Numerous studies have shown the efficacy of controlling individual cardiovascular risk factors in preventing or slowing ASCVD in people with diabetes. Furthermore, large benefits are seen when multiple cardiovascular risk factors are addressed simultaneously. Under the current paradigm of aggressive risk factor modification in people with diabetes, there is evidence that measures of 10-year CHD risk among U.S. adults with diabetes have improved significantly over the past decade (2) and that ASCVD morbidity and mortality have decreased (3,4).

Heart failure is another major cause of morbidity and mortality from cardiovascular disease. Recent studies have found that rates of incident heart failure hospitalization (adjusted for age and sex) were twofold higher in people with diabetes compared with those without (5,6). People with diabetes may have heart failure with preserved ejection fraction (HFpEF) or with reduced ejection fraction (HFrEF). Hypertension is often a precursor of heart failure of either type, and ASCVD can coexist with either type (7), whereas prior myocardial infarction (MI) is often a major factor in HFrEF. Rates of heart failure hospitalization have been improved in recent trials including people with type 2 diabetes, most of whom also had ASCVD, with sodium–glucose cotransporter 2 (SGLT2) inhibitors (811).

A recent meta-analysis indicated that SGLT2 inhibitors reduce the risk of heart failure hospitalization, cardiovascular mortality, and all-cause mortality in people with (secondary prevention) and without (primary prevention) cardiovascular disease (12).

For prevention and management of both ASCVD and heart failure, cardiovascular risk factors should be systematically assessed at least annually in all people with diabetes. These risk factors include duration of diabetes, obesity/overweight, hypertension, dyslipidemia, smoking, a family history of premature coronary disease, chronic kidney disease (CKD), and the presence of albuminuria. Modifiable abnormal risk factors should be treated as described in these guidelines. Notably, the majority of evidence supporting interventions to reduce cardiovascular risk in diabetes comes from trials of people with type 2 diabetes. No randomized trials have been specifically designed to assess the impact of cardiovascular risk reduction strategies in people with type 1 diabetes. Therefore, the recommendations for cardiovascular risk factor modification for people with type 1 diabetes are extrapolated from data obtained in people with type 2 diabetes and are similar to those for people with type 2 diabetes.

As depicted in Fig. 10.1, a comprehensive approach to the reduction in risk of diabetes-related complications is recommended. Therapy that includes multiple, concurrent evidence-based approaches to care will provide complementary reduction in the risks of microvascular, kidney, neurologic, and cardiovascular complications. Management of glycemia, blood pressure, and lipids and the incorporation of specific therapies with cardiovascular and kidney outcomes benefit (as individually appropriate) are considered fundamental elements of global risk reduction in diabetes.

Figure 10.1.

Figure 10.1

Multifactorial approach to reduction in risk of diabetes complications. *Risk reduction interventions to be applied as individually appropriate.

The Risk Calculator

The American College of Cardiology/American Heart Association ASCVD risk calculator (Risk Estimator Plus) is generally a useful tool to estimate 10-year risk of a first ASCVD event (available online at tools.acc.org/ASCVD-Risk-Estimator-Plus). The calculator includes diabetes as a risk factor, since diabetes itself confers increased risk for ASCVD, although it should be acknowledged that these risk calculators do not account for the duration of diabetes or the presence of diabetes complications, such as albuminuria. Although some variability in calibration exists in various subgroups, including by sex, race, and diabetes, the overall risk prediction does not differ in those with or without diabetes (1316), validating the use of risk calculators in people with diabetes. The 10-year risk of a first ASCVD event should be assessed to better stratify ASCVD risk and help guide therapy, as described below.

Recently, risk scores and other cardiovascular biomarkers have been developed for risk stratification of secondary prevention patients (i.e., those who are already high risk because they have ASCVD) but are not yet in widespread use (17,18). With newer, more expensive lipid-lowering therapies now available, use of these risk assessments may help target these new therapies to “higher risk” ASCVD patients in the future.

Hypertension/Blood Pressure Control

Hypertension is defined as a systolic blood pressure ≥130 mmHg or a diastolic blood pressure ≥80 mmHg (19). This is in agreement with the definition of hypertension by the American College of Cardiology and American Heart Association (19). Hypertension is common among people with either type 1 or type 2 diabetes. Hypertension is a major risk factor for both ASCVD and microvascular complications. Moreover, numerous studies have shown that antihypertensive therapy reduces ASCVD events, heart failure, and microvascular complications. Please refer to the American Diabetes Association position statement “Diabetes and Hypertension” for a detailed review of the epidemiology, diagnosis, and treatment of hypertension (20) and recent updated hypertension guideline recommendations (19,21,22).

Screening and Diagnosis

Recommendations

  • 10.1 Blood pressure should be measured at every routine clinical visit. When possible, individuals found to have elevated blood pressure (systolic blood pressure 120–129 mmHg and diastolic <80 mmHg) should have blood pressure confirmed using multiple readings, including measurements on a separate day, to diagnose hypertension. A Hypertension is defined as a systolic blood pressure ≥130 mmHg or a diastolic blood pressure ≥80 mmHg based on an average of ≥2 measurements obtained on ≥2 occasions. A Individuals with blood pressure ≥180/110 mmHg and cardiovascular disease could be diagnosed with hypertension at a single visit. E

  • 10.2 All people with hypertension and diabetes should monitor their blood pressure at home. A

Blood pressure should be measured at every routine clinical visit by a trained individual and should follow the guidelines established for the general population: measurement in the seated position, with feet on the floor and arm supported at heart level, after 5 min of rest. Cuff size should be appropriate for the upper-arm circumference. Elevated values should preferably be confirmed on a separate day; however, in individuals with cardiovascular disease and blood pressure ≥180/110 mmHg, it is reasonable to diagnose hypertension at a single visit (21). Postural changes in blood pressure and pulse may be evidence of autonomic neuropathy and therefore require adjustment of blood pressure targets. Orthostatic blood pressure measurements should be checked on initial visit and as indicated.

Home blood pressure self-monitoring and 24-h ambulatory blood pressure monitoring may provide evidence of white coat hypertension, masked hypertension, or other discrepancies between office and “true” blood pressure (23,24). In addition to confirming or refuting a diagnosis of hypertension, home blood pressure assessment may be useful to monitor antihypertensive treatment. Studies of individuals without diabetes found that home measurements may better correlate with ASCVD risk than office measurements (23,24). Moreover, home blood pressure monitoring may improve patient medication taking and thus help reduce cardiovascular risk (25).

Treatment Goals

Recommendations

  • 10.3 For people with diabetes and hypertension, blood pressure targets should be individualized through a shared decision-making process that addresses cardiovascular risk, potential adverse effects of antihypertensive medications, and patient preferences. B

  • 10.4 People with diabetes and hypertension qualify for antihypertensive drug therapy when the blood pressure is persistently elevated ≥130/80 mmHg. The on-treatment target blood pressure goal is <130/80 mmHg, if it can be safely attained. B

  • 10.5 In pregnant individuals with diabetes and chronic hypertension, a blood pressure threshold of 140/90 mmHg for initiation or titration of therapy is associated with better pregnancy outcomes than reserving treatment for severe hypertension, with no increase in risk of small-for-gestational age birth weight. A There are limited data on the optimal lower limit, but therapy should be lessened for blood pressure <90/60 mmHg. E A blood pressure target of 110–135/85 mmHg is suggested in the interest of reducing the risk for accelerated maternal hypertension. A

Randomized clinical trials have demonstrated unequivocally that treatment of hypertension reduces cardiovascular events as well as microvascular complications (2632). There has been controversy on the recommendation of a specific blood pressure goal in people with diabetes. The committee recognizes that there has been no randomized controlled trial to specifically demonstrate a decreased incidence of cardiovascular events in people with diabetes by targeting a blood pressure <130/80 mmHg. The recommendation to support a blood pressure goal of <130/80 mmHg in people with diabetes is consistent with guidelines from the American College of Cardiology and American Heart Association (20), the International Society of Hypertension (21), and the European Society of Cardiology (22). The committee’s recommendation for the blood pressure target of <130/80 mmHg derives primarily from the collective evidence of the following randomized controlled trials. The Systolic Blood Pressure Intervention Trial (SPRINT) demonstrated that treatment to a target systolic blood pressure of <120 mmHg decreases cardiovascular event rates by 25% in high-risk patients, although people with diabetes were excluded from this trial (33). The recently completed Strategy of Blood Pressure Intervention in the Elderly Hypertensive Patients (STEP) trial included nearly 20% of people with diabetes and noted decreased cardiovascular events with treatment of hypertension to a blood pressure target of <130 mmHg (34). While the ACCORD (Action to Control Cardiovascular Risk in Diabetes) blood pressure trial (ACCORD BP) did not confirm that targeting a systolic blood pressure of <120 mmHg in people with diabetes results in decreased cardiovascular event rates, the prespecified secondary outcome of stroke was reduced by 41% with intensive treatment (35). The Action in Diabetes and Vascular Disease: Preterax and Diamicron MR Controlled Evaluation (ADVANCE) trial revealed that treatment with perindopril/indapamide to an achieved systolic blood pressure of ∼135 mmHg significantly decreased cardiovascular event rates compared with a placebo treatment with an achieved blood pressure of 140 mmHg (36). Therefore, it is recommended that people with diabetes who have hypertension should be treated to blood pressure targets of <130/80 mmHg. Notably, there is an absence of high-quality data available to guide blood pressure targets in people with type 1 diabetes, but a similar blood pressure target of <130/80 mmHg is recommended in people with type 1 diabetes. As discussed below, treatment should be individualized and treatment should not be targeted to <120/80 mmHg, as a mean achieved blood pressure of <120/80 mmHg is associated with adverse events.

Randomized Controlled Trials of Intensive Versus Standard Blood Pressure Control

SPRINT provides the strongest evidence to support lower blood pressure goals in patients at increased cardiovascular risk, although this trial excluded people with diabetes (33). The trial enrolled 9,361 patients with a systolic blood pressure of ≥130 mmHg and increased cardiovascular risk and treated to a systolic blood pressure target of <120 mmHg (intensive treatment) versus a target of <140 mmHg (standard treatment). The primary composite outcome of myocardial infarction (MI), coronary syndromes, stroke, heart failure, or death from cardiovascular causes was reduced by 25% in the intensive treatment group. The achieved systolic blood pressures in the trial were 121 mmHg and 136 mmHg in the intensive versus standard treatment group, respectively. Adverse outcomes, including hypotension, syncope, electrolyte abnormality, and acute kidney injury were more common in the intensive treatment arm; risk of adverse outcomes needs to be weighed against the cardiovascular benefit of more intensive blood pressure lowering.

ACCORD BP provides the strongest direct assessment of the benefits and risks of intensive blood pressure control in people with type 2 diabetes (35). In the study, a total of 4,733 with type 2 diabetes were assigned to intensive therapy (targeting a systolic blood pressure <120 mmHg) or standard therapy (targeting a systolic blood pressure <140 mmHg). The mean achieved systolic blood pressures were 119 mmHg and 133 mmHg in the intensive versus standard group, respectively. The primary composite outcome of nonfatal MI, nonfatal stroke, or death from cardiovascular causes was not significantly reduced in the intensive treatment group. The prespecified secondary outcome of stroke was significantly reduced by 41% in the intensive treatment group. Adverse events attributed to blood pressure treatment, including hypotension, syncope, bradycardia, hyperkalemia, and elevations in serum creatinine occurred more frequently in the intensive treatment arm than in the standard therapy arm (Table 10.1).

Table 10.1.

Randomized controlled trials of intensive versus standard hypertension treatment strategies

Clinical trial Population Intensive Standard Outcomes
ACCORD BP (35) 4,733 participants with T2D aged 40–79 years with prior evidence of CVD or multiple cardiovascular risk factors SBP target: <120 mmHg
Achieved (mean) SBP/DBP: 119.3/64.4 mmHg
SBP target: 130–140 mmHg
Achieved (mean) SBP/DBP: 135/70.5 mmHg
• No benefit in primary end point: composite of nonfatal MI, nonfatal stroke, and CVD death
• Stroke risk reduced 41% with intensive control, not sustained through follow-up beyond the period of active treatment
• Adverse events more common in intensive group, particularly elevated serum creatinine and electrolyte abnormalities
ADVANCE (36) 11,140 participants with T2D aged ≥55 years with prior evidence of CVD or multiple cardiovascular risk factors Intervention: a single-pill, fixed-dose combination of perindopril and indapamide
Achieved (mean) SBP/DBP: 136/73 mmHg
Control: placebo
Achieved (mean) SBP/DBP: 141.6/75.2 mmHg
• Intervention reduced risk of primary composite end point of major macrovascular and microvascular events (9%), death from any cause (14%), and death from CVD (18%)
• 6-year observational follow-up found reduction in risk of death in intervention group attenuated but still significant (242)
HOT (37) 18,790 participants, including 1,501 with diabetes DBP target: ≤80 mmHg
Achieved (mean): 81.1 mmHg, ≤80 group; 85.2 mmHg, ≤90 group
DBP target: ≤90 mmHg • In the overall trial, there was no cardiovascular benefit with more intensive targets
• In the subpopulation with diabetes, an intensive DBP target was associated with a significantly reduced risk (51%) of CVD events
SPRINT (43) 9,361 participants without diabetes SBP target: <120 mmHg
Achieved (mean): 121.4 mmHg
SBP target: <140 mmHg
Achieved (mean): 136.2 mmHg
• Intensive SBP target lowered risk of the primary composite outcome 25% (MI, ACS, stroke, heart failure, and death due to CVD)
• Intensive target reduced risk of death 27%
• Intensive therapy increased risks of electrolyte abnormalities and AKI
STEP (34) 8,511 participants aged 60–80 years, including 1,627 with diabetes SBP target: <130 mmHg
Achieved (mean): 127.5 mmHg
SBP target: <150 mmHg
Achieved (mean): 135.3 mmHg
• Intensive SBP target lowered risk of the primary composite outcome 26% (stroke, ACS [acute MI and hospitalization for unstable angina], acute decompensated heart failure, coronary revascularization, atrial fibrillation, or death from cardiovascular causes)
• Intensive target reduced risk of cardiovascular death 28%
• Intensive therapy increased risks of hypotension

ACCORD BP, Action to Control Cardiovascular Risk in Diabetes Blood Pressure trial; ACS, acute coronary syndrome; ADVANCE, Action in Diabetes and Vascular Disease: Preterax and Diamicron MR Controlled Evaluation; AKI, acute kidney injury; CVD, cardiovascular disease; DBP, diastolic blood pressure; HOT, Hypertension Optimal Treatment trial; MI, myocardial infarction; SBP, systolic blood pressure; SPRINT, Systolic Blood Pressure Intervention Trial; STEP, Strategy of Blood Pressure Intervention in the Elderly Hypertensive Patients; T2D, type 2 diabetes.

Of note, the ACCORD BP and SPRINT trials targeted a similar systolic blood pressure <120 mmHg, but in contrast to SPRINT, the primary composite cardiovascular end point was nonsignificantly reduced in ACCORD BP. The results have been interpreted to be generally consistent between both trials, but ACCORD BP was viewed as underpowered due to the composite primary end point being less sensitive to blood pressure regulation (33).

The more recent STEP trial assigned 8,511 patients aged 60–80 years with hypertension to a systolic blood pressure target of 110 to <130 mmHg (intensive treatment) or a target of 130 to <150 mmHg (34). In this trial, the primary composite outcome of stroke, acute coronary syndrome, acute decompensated heart failure, coronary revascularization, atrial fibrillation, or death from cardiovascular causes was reduced by 26% in the intensive treatment group. In this trial, 18.9% of patients in the intensive treatment arm and 19.4% in the standard treatment arm had a diagnosis of type 2 diabetes. Hypotension occurred more frequently in the intensive treatment group (3.4%) compared with the standard treatment group (2.6%), without significant differences in other adverse events, including dizziness, syncope, or fractures.

In ADVANCE, 11,140 people with type 2 diabetes were randomized to receive either treatment with fixed combination perindopril/indapamide or matching placebo (36). The primary end point, a composite of cardiovascular death, nonfatal stroke infarction, or worsening renal or diabetic eye disease, was reduced by 9% in the combination treatment. The achieved systolic blood pressure was ∼135 mmHg in the treatment group and 140 mmHg in the placebo group.

The Hypertension Optimal Treatment (HOT) trial enrolled 18,790 patients and targeted diastolic blood pressure <90 mmHg, <85 mmHg, or <80 mmHg (37). The cardiovascular event rates, defined as fatal or nonfatal MI, fatal and nonfatal strokes, and all other cardiovascular events, were not significantly different between diastolic blood pressure targets (≤90 mmHg, ≤85 mmHg, and ≤80 mmHg), although the lowest incidence of cardiovascular events occurred with an achieved diastolic blood pressure of 82 mmHg. However, in people with diabetes, there was a significant 51% reduction in the treatment group with a target diastolic blood pressure of <80 mmHg compared with a target diastolic blood pressure of <90 mmHg.

Meta-analyses of Trials

To clarify optimal blood pressure targets in people with diabetes, multiple meta-analyses have been performed. One of the largest meta-analyses included 73,913 people with diabetes. Compared with a less tight blood pressure control, allocation to a tighter blood pressure control significantly reduced the risk of stroke by 31% but did not reduce the risk of MI (38). Another meta-analysis of 19 trials including 44,989 patients showed that a mean blood pressure of 133/76 mmHg is associated with a 14% risk reduction for major cardiovascular events compared with a mean blood pressure of 140/81 mmHg (32). This benefit was greatest in people with diabetes. An analysis of trials including people with type 2 diabetes and impaired glucose tolerance with achieved systolic blood pressures of <135 mmHg in the intensive blood pressure treatment group and <140 mmHg in the standard treatment group revealed a 10% reduction in all-cause mortality and a 17% reduction in stroke (30). More intensive reduction to <130 mmHg was associated with a further reduction in stroke but not other cardiovascular events.

Several meta-analyses stratified clinical trials by mean baseline blood pressure or mean blood pressure attained in the intervention (or intensive treatment) arm. Based on these analyses, antihypertensive treatment appears to be most beneficial when mean baseline blood pressure is ≥140/90 mmHg (19,26,27,2931). Among trials with lower baseline or attained blood pressure, antihypertensive treatment reduced the risk of stroke, retinopathy, and albuminuria, but effects on other ASCVD outcomes and heart failure were not evident.

Individualization of Treatment Targets

Patients and clinicians should engage in a shared decision-making process to determine individual blood pressure targets (19). This approach acknowledges that the benefits and risks of intensive blood pressure targets are uncertain and may vary across patients and is consistent with a patient-focused approach to care that values patient priorities and health care professional judgment (39). Secondary analyses of ACCORD BP and SPRINT suggest that clinical factors can help determine individuals more likely to benefit and less likely to be harmed by intensive blood pressure control (40,41).

Absolute benefit from blood pressure reduction correlated with absolute baseline cardiovascular risk in SPRINT and in earlier clinical trials conducted at higher baseline blood pressure levels (13,41). Extrapolation of these studies suggests that people with diabetes may also be more likely to benefit from intensive blood pressure control when they have high absolute cardiovascular risk. This approach is consistent with guidelines from the American College of Cardiology and American Heart Association, which also advocate a blood pressure target of <130/80 mmHg for all people, with or without diabetes (20).

Potential adverse effects of antihypertensive therapy (e.g., hypotension, syncope, falls, acute kidney injury, and electrolyte abnormalities) should also be taken into account (33,35,42,43). Individuals with older age, CKD, and frailty have been shown to be at higher risk of adverse effects of intensive blood pressure control (43). In addition, individuals with orthostatic hypotension, substantial comorbidity, functional limitations, or polypharmacy may be at high risk of adverse effects, and some patients may prefer higher blood pressure targets to enhance quality of life. However, in ACCORD BP, it was found that intensive blood pressure lowering decreased the risk of cardiovascular events irrespective of baseline diastolic blood pressure in patients who also received standard glycemic control (44). Therefore, the presence of low diastolic blood pressure is not necessarily a contraindication to more intensive blood pressure management in the context of otherwise standard care.

Pregnancy and Antihypertensive Medications

There are few randomized controlled trials of antihypertensive therapy in pregnant individuals with diabetes. A 2014 Cochrane systematic review of antihypertensive therapy for mild to moderate chronic hypertension that included 49 trials and over 4,700 women did not find any conclusive evidence for or against blood pressure treatment to reduce the risk of preeclampsia for the mother or effects on perinatal outcomes such as preterm birth, small-for-gestational-age infants, or fetal death (45). The Control of Hypertension in Pregnancy Study (CHIPS) (46) enrolled mostly women with chronic hypertension. In CHIPS, targeting a diastolic blood pressure of 85 mmHg during pregnancy was associated with reduced likelihood of developing accelerated maternal hypertension and no demonstrable adverse outcome for infants compared with targeting a higher diastolic blood pressure. The mean systolic blood pressure achieved in the more intensively treated group was 133.1 ± 0.5 mmHg, and the mean diastolic blood pressure achieved in that group was 85.3 ± 0.3 mmHg. A similar approach is supported by the International Society for the Study of Hypertension in Pregnancy, which specifically recommends use of antihypertensive therapy to maintain systolic blood pressure between 110 and 140 mmHg and diastolic blood pressure between 80 and 85 mmHg (47).

The more recent Chronic Hypertension and Pregnancy (CHAP) trial assigned pregnant individuals with mild chronic hypertension to antihypertensive medications to target a blood pressure goal of <140/90 mmHg (active treatment group) or to control treatment, in which antihypertensive therapy was withheld unless severe hypertension (systolic pressure ≥160 mmHg or diastolic pressure ≥105 mmHg) developed (control group) (48). The primary outcome, a composite of preeclampsia with severe features, medically indicated preterm birth at <35 weeks of gestation, placental abruption, or fetal/neonatal death, occurred in 30.2% of female participants in the active treatment group vs. 37.0% in the control group (P < 0.001). The mean systolic blood pressure between randomization and delivery was 129.5 mmHg in the active treatment group and 132.6 mmHg in the control group.

Current evidence supports controlling blood pressure to 110–135/85 mmHg to reduce the risk of accelerated maternal hypertension but also to minimize impairment of fetal growth. During pregnancy, treatment with ACE inhibitors, angiotensin receptor blockers (ARBs), and spironolactone are contraindicated as they may cause fetal damage. Special consideration should be taken for individuals of childbearing potential, and people intending to become pregnant should switch from an ACE inhibitor/ARB or spironolactone to an alternative antihypertensive medication approved during pregnancy. Antihypertensive drugs known to be effective and safe in pregnancy include methyldopa, labetalol, and long-acting nifedipine, while hydralzine may be considered in the acute management of hypertension in pregnancy or severe preeclampsia (49). Diuretics are not recommended for blood pressure control in pregnancy but may be used during late-stage pregnancy if needed for volume control (49,50). The American College of Obstetricians and Gynecologists also recommends that postpartum individuals with gestational hypertension, preeclampsia, and superimposed preeclampsia have their blood pressures observed for 72 h in the hospital and for 7–10 days postpartum. Long-term follow-up is recommended for these individuals as they have increased lifetime cardiovascular risk (51). See Section 15, “Management of Diabetes in Pregnancy,” for additional information.

Treatment Strategies

Lifestyle Intervention

Recommendation

  • 10.6 For people with blood pressure >120/80 mmHg, lifestyle intervention consists of weight loss when indicated, a Dietary Approaches to Stop Hypertension (DASH)-style eating pattern including reducing sodium and increasing potassium intake, moderation of alcohol intake, and increased physical activity. A

Lifestyle management is an important component of hypertension treatment because it lowers blood pressure, enhances the effectiveness of some antihypertensive medications, promotes other aspects of metabolic and vascular health, and generally leads to few adverse effects. Lifestyle therapy consists of reducing excess body weight through caloric restriction (see Section 8, “Obesity and Weight Management for the Prevention and Treatment of Type 2 Diabetes”), at least 150 min of moderate-intensity aerobic activity per week (see Section 3, “Prevention or Delay of Type 2 Diabetes and Associated Comorbidities”), restricting sodium intake (<2,300 mg/day), increasing consumption of fruits and vegetables (8–10 servings per day) and low-fat dairy products (2–3 servings per day), avoiding excessive alcohol consumption (no more than 2 servings per day in men and no more than 1 serving per day in women) (52), and increasing activity levels (53) (see Section 5, “Facilitating Positive Health Behaviors and Well-being to Improve Health Outcomes”).

These lifestyle interventions are reasonable for individuals with diabetes and mildly elevated blood pressure (systolic >120 mmHg or diastolic >80 mmHg) and should be initiated along with pharmacologic therapy when hypertension is diagnosed (Fig. 10.2) (53). A lifestyle therapy plan should be developed in collaboration with the patient and discussed as part of diabetes management. Use of internet or mobile-based digital platforms to reinforce healthy behaviors may be considered as a component of care, as these interventions have been found to enhance the efficacy of medical therapy for hypertension (54,55).

Figure 10.2.

Figure 10.2

Recommendations for the treatment of confirmed hypertension in people with diabetes. *An ACE inhibitor (ACEi) or angiotensin receptor blocker (ARB) is suggested to treat hypertension for people with coronary artery disease (CAD) or urine albumin-to-creatinine ratio 30–299 mg/g creatinine and strongly recommended for individuals with urine albumin-to-creatinine ratio ≥300 mg/g creatinine. **Thiazide-like diuretic; long-acting agents shown to reduce cardiovascular events, such as chlorthalidone and indapamide, are preferred. ***Dihydropyridine calcium channel blocker (CCB). BP, blood pressure. Adapted from de Boer et al. (20).

Pharmacologic Interventions

Recommendations

  • 10.7 Individuals with confirmed office-based blood pressure ≥130/80 mmHg qualify for initiation and titration of pharmacologic therapy to achieve the recommended blood pressure goal of <130/80 mmHg. A

  • 10.8 Individuals with confirmed office-based blood pressure ≥160/100 mmHg should, in addition to lifestyle therapy, have prompt initiation and timely titration of two drugs or a single-pill combination of drugs demonstrated to reduce cardiovascular events in people with diabetes. A

  • 10.9 Treatment for hypertension should include drug classes demonstrated to reduce cardiovascular events in people with diabetes. A ACE inhibitors or angiotensin receptor blockers are recommended first-line therapy for hypertension in people with diabetes and coronary artery disease. A

  • 10.10 Multiple-drug therapy is generally required to achieve blood pressure targets. However, combinations of ACE inhibitors and angiotensin receptor blockers and combinations of ACE inhibitors or angiotensin receptor blockers with direct renin inhibitors should not be used. A

  • 10.11 An ACE inhibitor or angiotensin receptor blocker, at the maximum tolerated dose indicated for blood pressure treatment, is the recommended first-line treatment for hypertension in people with diabetes and urinary albumin-to-creatinine ratio ≥300 mg/g creatinine A or 30–299 mg/g creatinine. B If one class is not tolerated, the other should be substituted. B

  • 10.12 For patients treated with an ACE inhibitor, angiotensin receptor blocker, or diuretic, serum creatinine/estimated glomerular filtration rate and serum potassium levels should be monitored at least annually. B

Initial Number of Antihypertensive Medications.

Initial treatment for people with diabetes depends on the severity of hypertension (Fig. 10.2). Those with blood pressure between 130/80 mmHg and 160/100 mmHg may begin with a single drug. For patients with blood pressure ≥160/100 mmHg, initial pharmacologic treatment with two antihypertensive medications is recommended in order to more effectively achieve adequate blood pressure control (5658). Single-pill antihypertensive combinations may improve medication taking in some patients (59).

Classes of Antihypertensive Medications.

Initial treatment for hypertension should include any of the drug classes demonstrated to reduce cardiovascular events in people with diabetes: ACE inhibitors (60,61), ARBs (60,61), thiazide-like diuretics (62), or dihydropyridine calcium channel blockers (63). In people with diabetes and established coronary artery disease, ACE inhibitors or ARBs are recommended first-line therapy for hypertension (6466). For patients with albuminuria (urine albumin-to-creatinine ratio [UACR] ≥30 mg/g), initial treatment should include an ACE inhibitor or ARB to reduce the risk of progressive kidney disease (20) (Fig. 10.2). In patients receiving ACE inhibitor or ARB therapy, continuation of those medications as kidney function declines to estimated glomerular filtration rate (eGFR) <30 mL/min/1.73 m2 may provide cardiovascular benefit without significantly increasing the risk of end-stage kidney disease (67). In the absence of albuminuria, risk of progressive kidney disease is low, and ACE inhibitors and ARBs have not been found to afford superior cardioprotection when compared with thiazide-like diuretics or dihydropyridine calcium channel blockers (68). β-Blockers are indicated in the setting of prior MI, active angina, or HfrEF but have not been shown to reduce mortality as blood pressure-lowering agents in the absence of these conditions (28,69,70).

Multiple-Drug Therapy.

Multiple-drug therapy is often required to achieve blood pressure targets (Fig. 10.2), particularly in the setting of diabetic kidney disease. However, the use of both ACE inhibitors and ARBs in combination, or the combination of an ACE inhibitor or ARB and a direct renin inhibitor, is contraindicated given the lack of added ASCVD benefit and increased rate of adverse events—namely, hyperkalemia, syncope, and acute kidney injury (AKI) (7173). Titration of and/or addition of further blood pressure medications should be made in a timely fashion to overcome therapeutic inertia in achieving blood pressure targets.

Bedtime Dosing.

Although prior analyses of randomized clinical trials found a benefit to evening versus morning dosing of antihypertensive medications (74,75), these results have not been reproduced in subsequent trials. Therefore, preferential use of antihypertensives at bedtime is not recommended (76).

Hyperkalemia and Acute Kidney Injury.

Treatment with ACE inhibitors or ARBs can cause AKI and hyperkalemia, while diuretics can cause AKI and either hypokalemia or hyperkalemia (depending on mechanism of action) (77,78). Detection and management of these abnormalities is important because AKI and hyperkalemia each increase the risks of cardiovascular events and death (79). Therefore, serum creatinine and potassium should be monitored during treatment with an ACE inhibitor, ARB, or diuretic, particularly among patients with reduced glomerular filtration who are at increased risk of hyperkalemia and AKI (77,78,80).

Resistant Hypertension

Recommendation

  • 10.13 Individuals with hypertension who are not meeting blood pressure targets on three classes of antihypertensive medications (including a diuretic) should be considered for mineralocorticoid receptor antagonist therapy. A

Resistant hypertension is defined as blood pressure ≥140/90 mmHg despite a therapeutic strategy that includes appropriate lifestyle management plus a diuretic and two other antihypertensive drugs with complementary mechanisms of action at adequate doses. Prior to diagnosing resistant hypertension, a number of other conditions should be excluded, including missed doses of antihypertensive medications, white coat hypertension, and secondary hypertension. In general, barriers to medication taking (such as cost and side effects) should be identified and addressed (Fig. 10.2). Mineralocorticoid receptor antagonists, including spironolactone and eplerenone, are effective for management of resistant hypertension in people with type 2 diabetes when added to existing treatment with an ACE inhibitor or ARB, thiazide-like diuretic, or dihydropyridine calcium channel blocker (81). In addition, mineralocorticoid receptor antagonists reduce albuminuria in people with diabetic nephropathy (8284). However, adding a mineralocorticoid receptor antagonist to a regimen including an ACE inhibitor or ARB may increase the risk for hyperkalemia, emphasizing the importance of regular monitoring for serum creatinine and potassium in these patients, and long-term outcome studies are needed to better evaluate the role of mineralocorticoid receptor antagonists in blood pressure management.

Lipid Management

Lifestyle Intervention

Recommendations

  • 10.14 Lifestyle modification focusing on weight loss (if indicated); application of a Mediterranean or Dietary Approaches to Stop Hypertension (DASH) eating pattern; reduction of saturated fat and trans fat; increase of dietary n-3 fatty acids, viscous fiber, and plant stanols/sterols intake; and increased physical activity should be recommended to improve the lipid profile and reduce the risk of developing atherosclerotic cardiovascular disease in people with diabetes. A

  • 10.15 Intensify lifestyle therapy and optimize glycemic control for patients with elevated triglyceride levels (≥150 mg/dL [1.7 mmol/L]) and/or low HDL cholesterol (<40 mg/dL [1.0 mmol/L] for men, <50 mg/dL [1.3 mmol/L] for women). C

Lifestyle intervention, including weight loss in people with overweight or obesity (when appropriate) (85), increased physical activity, and medical nutrition therapy, allows some patients to reduce ASCVD risk factors. Nutrition intervention should be tailored according to each patient’s age, pharmacologic treatment, lipid levels, and medical conditions.

Recommendations should focus on application of a Mediterranean (83) or Dietary Approaches to Stop Hypertension (DASH) eating pattern, reducing saturated and trans fat intake and increasing plant stanols/sterols, n-3 fatty acids, and viscous fiber (such as in oats, legumes, and citrus) intake (86,87). Glycemic control may also beneficially modify plasma lipid levels, particularly in patients with very high triglycerides and poor glycemic control. See Section 5, “Facilitating Positive Health Behaviors and Well-being to Improve Health Outcomes,” for additional nutrition information.

Ongoing Therapy and Monitoring with Lipid Panel

Recommendations

  • 10.16 In adults not taking statins or other lipid-lowering therapy, it is reasonable to obtain a lipid profile at the time of diabetes diagnosis, at an initial medical evaluation, and every 5 years thereafter if under the age of 40 years, or more frequently if indicated. E

  • 10.17 Obtain a lipid profile at initiation of statins or other lipid-lowering therapy, 4–12 weeks after initiation or a change in dose, and annually thereafter as it may help to monitor the response to therapy and inform medication taking. E

In adults with diabetes, it is reasonable to obtain a lipid profile (total cholesterol, LDL cholesterol, HDL cholesterol, and triglycerides) at the time of diagnosis, at the initial medical evaluation, and at least every 5 years thereafter in patients <40 years of age. In younger people with longer duration of disease (such as those with youth-onset type 1 diabetes), more frequent lipid profiles may be reasonable. A lipid panel should also be obtained immediately before initiating statin therapy. Once a patient is taking a statin, LDL cholesterol levels should be assessed 4–12 weeks after initiation of statin therapy, after any change in dose, and on an individual basis (e.g., to monitor for medication taking and efficacy). If LDL cholesterol levels are not responding in spite of medication taking, clinical judgment is recommended to determine the need for and timing of lipid panels. In individual patients, the highly variable LDL cholesterol–lowering response seen with statins is poorly understood (88). Clinicians should attempt to find a dose or alternative statin that is tolerable if side effects occur. There is evidence for benefit from even extremely low, less than daily statin doses (89).

Statin Treatment

Primary Prevention

Recommendations

  • 10.18 For people with diabetes aged 40–75 years without atherosclerotic cardiovascular disease, use moderate-intensity statin therapy in addition to lifestyle therapy. A

  • 10.19 For people with diabetes aged 20–39 years with additional atherosclerotic cardiovascular disease risk factors, it may be reasonable to initiate statin therapy in addition to lifestyle therapy. C

  • 10.20 For people with diabetes aged 40–75 at higher cardiovascular risk, including those with one or more atherosclerotic cardiovascular disease risk factors, it is recommended to use high-intensity statin therapy to reduce LDL cholesterol by ≥50% of baseline and to target an LDL cholesterol goal of <70 mg/dL. B

  • 10.21 For people with diabetes aged 40–75 years at higher cardiovascular risk, especially those with multiple atherosclerotic cardiovascular disease risk factors and an LDL cholesterol ≥70 mg/dL, it may be reasonable to add ezetimibe or a PCSK9 inhibitor to maximum tolerated statin therapy. C

  • 10.22 In adults with diabetes aged >75 years already on statin therapy, it is reasonable to continue statin treatment. B

  • 10.23 In adults with diabetes aged >75 years, it may be reasonable to initiate moderate-intensity statin therapy after discussion of potential benefits and risks. C

  • 10.24 Statin therapy is contraindicated in pregnancy. B

Secondary Prevention

Recommendations

  • 10.25 For people of all ages with diabetes and atherosclerotic cardiovascular disease, high-intensity statin therapy should be added to lifestyle therapy. A

  • 10.26 For people with diabetes and atherosclerotic cardiovascular disease, treatment with high-intensity statin therapy is recommended to target an LDL cholesterol reduction of ≥50% from baseline and an LDL cholesterol goal of <55 mg/dL. Addition of ezetimibe or a PCSK9 inhibitor with proven benefit in this population is recommended if this goal is not achieved on maximum tolerated statin therapy. B

  • 10.27 For individuals who do not tolerate the intended intensity, the maximum tolerated statin dose should be used. E

Initiating Statin Therapy Based on Risk

People with type 2 diabetes have an increased prevalence of lipid abnormalities, contributing to their high risk of ASCVD. Multiple clinical trials have demonstrated the beneficial effects of statin therapy on ASCVD outcomes in subjects with and without CHD (90,91). Subgroup analyses of people with diabetes in larger trials (9296) and trials in people with diabetes (97,98) showed significant primary and secondary prevention of ASCVD events and CHD death in people with diabetes. Meta-analyses, including data from over 18,000 people with diabetes from 14 randomized trials of statin therapy (mean follow-up 4.3 years), demonstrate a 9% proportional reduction in all-cause mortality and 13% reduction in vascular mortality for each 1 mmol/L (39 mg/dL) reduction in LDL cholesterol (99). The cardiovascular benefit in this large meta-analysis did not depend on baseline LDL cholesterol levels and was linearly related to the LDL cholesterol reduction without a low threshold beyond which there was no benefit observed (99).

Accordingly, statins are the drugs of choice for LDL cholesterol lowering and cardioprotection. Table 10.2 shows the two statin dosing intensities that are recommended for use in clinical practice: high-intensity statin therapy will achieve approximately a ≥50% reduction in LDL cholesterol, and moderate-intensity statin regimens achieve 30–49% reductions in LDL cholesterol. Low-dose statin therapy is generally not recommended in people with diabetes but is sometimes the only dose of statin that a patient can tolerate. For patients who do not tolerate the intended intensity of statin, the maximum tolerated statin dose should be used.

Table 10.2.

High-intensity and moderate-intensity statin therapy*

High-intensity statin therapy (lowers LDL cholesterol by ≥50%) Moderate-intensity statin therapy (lowers LDL cholesterol by 30–49%)
Atorvastatin 40–80 mg Atorvastatin 10–20 mg
Rosuvastatin 20–40 mg Rosuvastatin 5–10 mg
Simvastatin 20–40 mg
Pravastatin 40–80 mg
Lovastatin 40 mg
Fluvastatin XL 80 mg
Pitavastatin 1–4 mg
*

Once-daily dosing. XL, extended release.

As in those without diabetes, absolute reductions in ASCVD outcomes (CHD death and nonfatal MI) are greatest in people with high baseline ASCVD risk (known ASCVD and/or very high LDL cholesterol levels), but the overall benefits of statin therapy in people with diabetes at moderate or even low risk for ASCVD are convincing (100,101). The relative benefit of lipid-lowering therapy has been uniform across most subgroups tested (91,99), including subgroups that varied with respect to age and other risk factors.

Primary Prevention (People without ASCVD)

For primary prevention, moderate-dose statin therapy is recommended for those aged ≥40 years (93,100,101), although high-intensity therapy should be considered in the context of additional ASCVD risk factors. The evidence is strong for people with diabetes aged 40–75 years, an age-group well represented in statin trials showing benefit. Since cardiovascular risk is enhanced in people with diabetes, as noted above, patients who also have multiple other coronary risk factors have increased risk, equivalent to that of those with ASCVD. Therefore, current guidelines recommend that in people with diabetes who are at higher cardiovascular risk, especially those with one or more ASCVD risk factors, high-intensity statin therapy should be prescribed to reduce LDL cholesterol by ≥50% from baseline and to target an LDL cholesterol of <70 mg/dL (102104). Since in clinical practice it is frequently difficult to ascertain the baseline LDL cholesterol level prior to statin therapy initiation, in those individuals, a focus on an LDL cholesterol target level of <70 mg/dL rather than the percent reduction in LDL cholesterol is recommended. In those individuals, it may also be reasonable to add ezetimibe or proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor therapy to maximum tolerated statin therapy if needed to reduce LDL cholesterol levels by ≥50% and to achieve the recommended LDL cholesterol target of <70 mg/dL (14). The evidence is lower for patients aged >75 years; relatively few older people with diabetes have been enrolled in primary prevention trials. However, heterogeneity by age has not been seen in the relative benefit of lipid-lowering therapy in trials that included older participants (91,98,99), and because older age confers higher risk, the absolute benefits are actually greater (91,105). Moderate-intensity statin therapy is recommended in people with diabetes who are ≥75 years of age. However, the risk-benefit profile should be routinely evaluated in this population, with downward titration of dose performed as needed. See Section 13, “Older Adults,” for more details on clinical considerations for this population.

Age <40 Years and/or Type 1 Diabetes.

Very little clinical trial evidence exists for people with type 2 diabetes under the age of 40 years or for people with type diabetes of any age. For pediatric recommendations, see Section 14, “Children and Adolescents.” In the Heart Protection Study (lower age limit 40 years), the subgroup of ∼600 people with type 1 diabetes had a proportionately similar, although not statistically significant, reduction in risk to that in people with type 2 diabetes (93). Even though the data are not definitive, similar statin treatment approaches should be considered for people with type 1 or type 2 diabetes, particularly in the presence of other cardiovascular risk factors. Patients <40 years of age have lower risk of developing a cardiovascular event over a 10-year horizon; however, their lifetime risk of developing cardiovascular disease and suffering an MI, stroke, or cardiovascular death is high. For people who are <40 years of age and/or have type 1 diabetes with other ASCVD risk factors, it is recommended that the patient and health care professional discuss the relative benefits and risks and consider the use of moderate-intensity statin therapy. Please refer to “Type 1 Diabetes Mellitus and Cardiovascular Disease: A Scientific Statement From the American Heart Association and American Diabetes Association” (106) for additional discussion.

Secondary Prevention (People with ASCVD)

Because cardiovascular event rates are increased in people with diabetes and established ASCVD, intensive therapy is indicated and has been shown to be of benefit in multiple large meta-analyses and randomized cardiovascular outcomes trials (91,99,105,107,108). High-intensity statin therapy is recommended for all people with diabetes and ASCVD to target an LDL cholesterol reduction of ≥50% from baseline and an LDL cholesterol goal of <55 mg/dL. Based on the evidence discussed below, addition of ezetimibe or a PCSK9 inhibitor is recommended if this goal is not achieved on maximum tolerated statin therapy. These recommendations are based on the observation that high-intensity versus moderate-intensity statin therapy reduces cardiovascular event rates in high-risk individuals with established cardiovascular disease in randomized trials (95,107). In addition, the Cholesterol Treatment Trialists’ Collaboration involving 26 statin trials, of which 5 compared high-intensity versus moderate-intensity statins (99), showed a 21% reduction in major cardiovascular events in people with diabetes for every 39 mg/dL of LDL cholesterol lowering, irrespective of baseline LDL cholesterol or patient characteristics (99). However, the best evidence to support lower LDL cholesterol targets in people with diabetes and established cardiovascular disease derives from multiple large randomized trials investigating the benefits of adding nonstatin agents to statin therapy. As discussed in detail below, these include combination treatment with statins and ezetimibe (105,109) or PCSK9 inhibitors (108,110112). Each trial found a significant benefit in the reduction of ASCVD events that was directly related to the degree of further LDL cholesterol lowering. These large trials included a significant number of participants with diabetes and prespecified analyses on cardiovascular outcomes in people with and without diabetes (109,111,112). The decision to add a nonstatin agent should be made following a clinician-patient discussion about the net benefit, safety, and cost of combination therapy.

Combination Therapy for LDL Cholesterol Lowering

Statins and Ezetimibe

The IMProved Reduction of Outcomes: Vytorin Efficacy International Trial (IMPROVE-IT) was a randomized controlled trial in 18,144 patients comparing the addition of ezetimibe to simvastatin therapy versus simvastatin alone (105). Individuals were ≥50 years of age, had experienced a recent acute coronary syndrome (ACS) and were treated for an average of 6 years. Overall, the addition of ezetimibe led to a 6.4% relative benefit and a 2% absolute reduction in major adverse cardiovascular events (atherosclerotic cardiovascular events), with the degree of benefit being directly proportional to the change in LDL cholesterol, which was 70 mg/dL in the statin group on average and 54 mg/dL in the combination group (105). In those with diabetes (27% of participants), the combination of moderate-intensity simvastatin (40 mg) and ezetimibe (10 mg) showed a significant reduction of major adverse cardiovascular events with an absolute risk reduction of 5% (40% vs. 45% cumulative incidence at 7 years) and a relative risk reduction of 14% (hazard ratio [HR] 0.86 [95% CI 0.78–0.94]) over moderate-intensity simvastatin (40 mg) alone (109).

Statins and PCSK9 Inhibitors

Placebo-controlled trials evaluating the addition of the PCSK9 inhibitors evolocumab and alirocumab to maximum tolerated doses of statin therapy in participants who were at high risk for ASCVD demonstrated an average reduction in LDL cholesterol ranging from 36 to 59%. These agents have been approved as adjunctive therapy for individuals with ASCVD or familial hypercholesterolemia who are receiving maximum tolerated statin therapy but require additional lowering of LDL cholesterol (113,114). No cardiovascular outcome trials have been performed to assess whether PCSK9 inhibitor therapy reduces ASCVD event rates in individuals without established cardiovascular disease (primary prevention).

The effects of PCSK9 inhibition on ASCVD outcomes was investigated in the Further Cardiovascular Outcomes Research With PCSK9 Inhibition in Subjects With Elevated Risk (FOURIER) trial, which enrolled 27,564 individuals with prior ASCVD and an additional high-risk feature who were receiving their maximum tolerated statin therapy (two-thirds were on high-intensity statin) but who still had LDL cholesterol ≥70 mg/dL or non-HDL cholesterol ≥100 mg/dL (108). Patients were randomized to receive subcutaneous injections of evolocumab (either 140 mg every 2 weeks or 420 mg every month based on patient preference) versus placebo. Evolocumab reduced LDL cholesterol by 59% from a median of 92 to 30 mg/dL in the treatment arm.

During the median follow-up of 2.2 years, the composite outcome of cardiovascular death, MI, stroke, hospitalization for angina, or revascularization occurred in 11.3% vs. 9.8% of the placebo and evolocumab groups, respectively, representing a 15% relative risk reduction (P < 0.001). The combined end point of cardiovascular death, MI, or stroke was reduced by 20%, from 7.4 to 5.9% (P < 0.001). Evolocumab therapy also significantly reduced all strokes (1.5% vs. 1.9%; HR 0.79 [95% CI 0.66–0.95]; P = 0.01) and ischemic stroke (1.2% vs. 1.6%; HR 0.75 [95% CI 0.62–0.92]; P = 0.005) in the total population, with findings being consistent in individuals with or without a history of ischemic stroke at baseline (115). Importantly, similar benefits were seen in a prespecified subgroup of people with diabetes, comprising 11,031 patients (40% of the trial) (112).

In the ODYSSEY OUTCOMES trial (Evaluation of Cardiovascular Outcomes After an Acute Coronary Syndrome During Treatment With Alirocumab), 18,924 patients (28.8% of whom had diabetes) with recent acute coronary syndrome were randomized to the PCSK9 inhibitor alirocumab or placebo every 2 weeks in addition to maximum tolerated statin therapy, with alirocumab dosing titrated between 75 and 150 mg to achieve LDL cholesterol levels between 25 and 50 mg/dL (110). Over a median follow-up of 2.8 years, a composite primary end point (comprising death from CHD, nonfatal MI, fatal or nonfatal ischemic stroke, or unstable angina requiring hospital admission) occurred in 903 patients (9.5%) in the alirocumab group and in 1,052 patients (11.1%) in the placebo group (HR 0.85 [95% CI 0.78–0.93]; P < 0.001). Combination therapy with alirocumab plus statin therapy resulted in a greater absolute reduction in the incidence of the primary end point in people with diabetes (2.3% [95% CI 0.4–4.2]) than in those with prediabetes (1.2% [0.0–2.4]) or normoglycemia (1.2% [–0.3 to 2.7]) (111).

In addition to monoclonal antibodies targeting PCSK9, the siRNA inclisiran has been developed and has recently become available in the U.S. In the Inclisiran for Participants With Atherosclerotic Cardiovascular Disease and Elevated Low-density Lipoprotein Cholesterol (ORION-10) and Inclisiran for Subjects With ASCVD or ASCVD-Risk Equivalents and Elevated Low-density Lipoprotein Cholesterol (ORION-11) trials (116), individuals with established cardiovascular disease or ASCVD risk equivalent were randomized to receive inclisiran or placebo. Inclisiran allows less frequent administration compared with monoclonal antibodies and was administered on day 1, on day 90, and every 6 months in these trials. In the ORION-10 trial, 47.5% of patients in the inclisiran group and 42.4% in the placebo group had diabetes; in the ORION-11 trial, 36.5% of patients in the inclisiran group and 33.7% in the placebo group had diabetes. The coprimary end point of placebo-corrected percentage change in LDL cholesterol level from baseline to day 510 was 52.3% in the ORION-10 trial and 49.9% in the ORION-11 trial. In an exploratory analysis, the prespecified cardiovascular end point, defined as a cardiovascular basket of nonadjudicated terms, including those classified within cardiac death, and any signs or symptoms of cardiac arrest, nonfatal MI, or stroke, occurred in 7.4% of the inclisiran group and 10.2% of the placebo group in the ORION-10 trial and in 7.8% of the inclisiran group and 10.3% of the placebo group in the ORION-11 trial. A cardiovascular outcome trial using inclisiran in people with established cardiovascular disease is currently ongoing (117).

Statins and Bempedoic Acid

Bempedoic acid is a novel LDL cholesterol–lowering agent that is indicated as an adjunct to diet and maximum tolerated statin therapy for the treatment of adults with heterozygous familial hypercholesterolemia or established ASCVD who require additional lowering of LDL cholesterol. A pooled analysis suggests that bempedoic acid therapy lowers LDL cholesterol levels by about 23% compared with placebo (118). At this time, there are no completed trials demonstrating a cardiovascular outcomes benefit to use of this medication; however, this agent may be considered for patients who cannot use or tolerate other evidence-based LDL cholesterol-lowering approaches, or for whom those other therapies are inadequately effective (119).

Treatment of Other Lipoprotein Fractions or Targets

Recommendations

  • 10.28 For individuals with fasting triglyceride levels ≥500 mg/dL, evaluate for secondary causes of hypertriglyceridemia and consider medical therapy to reduce the risk of pancreatitis. C

  • 10.29 In adults with moderate hypertriglyceridemia (fasting or nonfasting triglycerides 175–499 mg/dL), clinicians should address and treat lifestyle factors (obesity and metabolic syndrome), secondary factors (diabetes, chronic liver or kidney disease and/or nephrotic syndrome, hypothyroidism), and medications that raise triglycerides. C

  • 10.30 In individuals with atherosclerotic cardiovascular disease or other cardiovascular risk factors on a statin with controlled LDL cholesterol but elevated triglycerides (135–499 mg/dL), the addition of icosapent ethyl can be considered to reduce cardiovascular risk. A

Hypertriglyceridemia should be addressed with dietary and lifestyle changes including weight loss and abstinence from alcohol (120). Severe hypertriglyceridemia (fasting triglycerides ≥500 mg/dL and especially >1,000 mg/dL) may warrant pharmacologic therapy (fibric acid derivatives and/or fish oil) and reduction in dietary fat to reduce the risk of acute pancreatitis. Moderate- or high-intensity statin therapy should also be used as indicated to reduce risk of cardiovascular events (see statin treatment). In people with moderate hypertriglyceridemia, lifestyle interventions, treatment of secondary factors, and avoidance of medications that might raise triglycerides are recommended.

The Reduction of Cardiovascular Events with Icosapent Ethyl-Intervention Trial (REDUCE-IT) enrolled 8,179 adults receiving statin therapy with moderately elevated triglycerides (135–499 mg/dL, median baseline of 216 mg/dL) who had either established cardiovascular disease (secondary prevention cohort) or diabetes plus at least one other cardiovascular risk factor (primary prevention cohort) (121). Patients were randomized to icosapent ethyl 4 g/day (2 g twice daily with food) versus placebo. The trial met its primary end point, demonstrating a 25% relative risk reduction (P < 0.001) for the primary end point composite of cardiovascular death, nonfatal MI, nonfatal stroke, coronary revascularization, or unstable angina. This reduction in risk was seen in people with or without diabetes at baseline. The composite of cardiovascular death, nonfatal MI, or nonfatal stroke was reduced by 26% (P < 0.001). Additional ischemic end points were significantly lower in the icosapent ethyl group than in the placebo group, including cardiovascular death, which was reduced by 20% (P = 0.03). The proportions of patients experiencing adverse events and serious adverse events were similar between the active and placebo treatment groups. It should be noted that data are lacking with other n-3 fatty acids, and results of the REDUCE-IT trial should not be extrapolated to other products (121). As an example, the addition of 4 g per day of a carboxylic acid formulation of the n-3 fatty acids eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) (n-3 carboxylic acid) to statin therapy in patients with atherogenic dyslipidemia and high cardiovascular risk, 70% of whom had diabetes, did not reduce the risk of major adverse cardiovascular events compared with the inert comparator of corn oil (122).

Low levels of HDL cholesterol, often associated with elevated triglyceride levels, are the most prevalent pattern of dyslipidemia in people with type 2 diabetes. However, the evidence for the use of drugs that target these lipid fractions is substantially less robust than that for statin therapy (123). In a large trial in people with diabetes, fenofibrate failed to reduce overall cardiovascular outcomes (124).

Other Combination Therapy

Recommendations

  • 10.31 Statin plus fibrate combination therapy has not been shown to improve atherosclerotic cardiovascular disease outcomes and is generally not recommended. A

  • 10.32 Statin plus niacin combination therapy has not been shown to provide additional cardiovascular benefit above statin therapy alone, may increase the risk of stroke with additional side effects, and is generally not recommended. A

Statin and Fibrate Combination Therapy

Combination therapy (statin and fibrate) is associated with an increased risk for abnormal transaminase levels, myositis, and rhabdomyolysis. The risk of rhabdomyolysis is more common with higher doses of statins and renal insufficiency and appears to be higher when statins are combined with gemfibrozil (compared with fenofibrate) (125).

In the ACCORD study, in people with type 2 diabetes who were at high risk for ASCVD, the combination of fenofibrate and simvastatin did not reduce the rate of fatal cardiovascular events, nonfatal MI, or nonfatal stroke compared with simvastatin alone. Prespecified subgroup analyses suggested heterogeneity in treatment effects with possible benefit for men with both a triglyceride level ≥204 mg/dL (2.3 mmol/L) and an HDL cholesterol level ≤34 mg/dL (0.9 mmol/L) (126).

Statin and Niacin Combination Therapy

The Atherothrombosis Intervention in Metabolic Syndrome With Low HDL/High Triglycerides: Impact on Global Health Outcomes (AIM-HIGH) trial randomized over 3,000 people (about one-third with diabetes) with established ASCVD, LDL cholesterol levels <180 mg/dL [4.7 mmol/L], low HDL cholesterol levels (men <40 mg/dL [1.0 mmol/L] and women <50 mg/dL [1.3 mmol/L]), and triglyceride levels of 150–400 mg/dL (1.7–4.5 mmol/L) to statin therapy plus extended-release niacin or placebo. The trial was halted early due to lack of efficacy on the primary ASCVD outcome (first event of the composite of death from CHD, nonfatal MI, ischemic stroke, hospitalization for an ACS, or symptom-driven coronary or cerebral revascularization) and a possible increase in ischemic stroke in those on combination therapy (127).

The much larger Heart Protection Study 2–Treatment of HDL to Reduce the Incidence of Vascular Events (HPS2-THRIVE) trial also failed to show a benefit of adding niacin to background statin therapy (128). A total of 25,673 individuals with prior vascular disease were randomized to receive 2 g of extended-release niacin and 40 mg of laropiprant (an antagonist of the prostaglandin D2 receptor DP1 that has been shown to improve participation in niacin therapy) versus a matching placebo daily and followed for a median follow-up period of 3.9 years. There was no significant difference in the rate of coronary death, MI, stroke, or coronary revascularization with the addition of niacin–laropiprant versus placebo (13.2% vs. 13.7%; rate ratio 0.96; P = 0.29). Niacin–laropiprant was associated with an increased incidence of new-onset diabetes (absolute excess, 1.3 percentage points; P < 0.001) and disturbances in diabetes management among those with diabetes. In addition, there was an increase in serious adverse events associated with the gastrointestinal system, musculoskeletal system, skin, and, unexpectedly, infection and bleeding.

Therefore, combination therapy with a statin and niacin is not recommended given the lack of efficacy on major ASCVD outcomes and increased side effects.

Diabetes Risk with Statin Use

Several studies have reported a modestly increased risk of incident diabetes with statin use (129,130), which may be limited to those with diabetes risk factors. An analysis of one of the initial studies suggested that although statin use was associated with diabetes risk, the cardiovascular event rate reduction with statins far outweighed the risk of incident diabetes even for patients at highest risk for diabetes (131). The absolute risk increase was small (over 5 years of follow-up, 1.2% of participants on placebo developed diabetes and 1.5% on rosuvastatin developed diabetes) (131). A meta-analysis of 13 randomized statin trials with 91,140 participants showed an odds ratio of 1.09 for a new diagnosis of diabetes, so that (on average) treatment of 255 patients with statins for 4 years resulted in one additional case of diabetes while simultaneously preventing 5.4 vascular events among those 255 patients (130).

Lipid-Lowering Agents and Cognitive Function

Although concerns regarding a potential adverse impact of lipid-lowering agents on cognitive function have been raised, several lines of evidence point against this association, as detailed in a 2018 European Atherosclerosis Society Consensus Panel statement (132). First, there are three large randomized trials of statin versus placebo where specific cognitive tests were performed, and no differences were seen between statin and placebo (133136). In addition, no change in cognitive function has been reported in studies with the addition of ezetimibe (105) or PCSK9 inhibitors (108,137) to statin therapy, including among patients treated to very low LDL cholesterol levels. In addition, the most recent systematic review of the U.S. Food and Drug Administration’s (FDA’s) postmarketing surveillance databases, randomized controlled trials, and cohort, case-control, and cross-sectional studies evaluating cognition in patients receiving statins found that published data do not reveal an adverse effect of statins on cognition (138). Therefore, a concern that statins or other lipid-lowering agents might cause cognitive dysfunction or dementia is not currently supported by evidence and should not deter their use in individuals with diabetes at high risk for ASCVD (138).

Antiplatelet Agents

Recommendations

  • 10.33 Use aspirin therapy (75–162 mg/day) as a secondary prevention strategy in those with diabetes and a history of atherosclerotic cardiovascular disease. A

  • 10.34 For individuals with atherosclerotic cardiovascular disease and documented aspirin allergy, clopidogrel (75 mg/day) should be used. B

  • 10.35 Dual antiplatelet therapy (with low-dose aspirin and a P2Y12 inhibitor) is reasonable for a year after an acute coronary syndrome and may have benefits beyond this period. A

  • 10.36 Long-term treatment with dual antiplatelet therapy should be considered for individuals with prior coronary intervention, high ischemic risk, and low bleeding risk to prevent major adverse cardiovascular events. A

  • 10.37 Combination therapy with aspirin plus low-dose rivaroxaban should be considered for individuals with stable coronary and/or peripheral artery disease and low bleeding risk to prevent major adverse limb and cardiovascular events. A

  • 10.38 Aspirin therapy (75–162 mg/day) may be considered as a primary prevention strategy in those with diabetes who are at increased cardiovascular risk, after a comprehensive discussion with the patient on the benefits versus the comparable increased risk of bleeding. A

Risk Reduction

Aspirin has been shown to be effective in reducing cardiovascular morbidity and mortality in high-risk patients with previous MI or stroke (secondary prevention) and is strongly recommended. In primary prevention, however, among patients with no previous cardiovascular events, its net benefit is more controversial (129,140).

Previous randomized controlled trials of aspirin specifically in people with diabetes failed to consistently show a significant reduction in overall ASCVD end points, raising questions about the efficacy of aspirin for primary prevention in people with diabetes, although some sex differences were suggested (141143).

The Antithrombotic Trialists’ Collaboration published an individual patient–level meta-analysis (139) of the six large trials of aspirin for primary prevention in the general population. These trials collectively enrolled over 95,000 participants, including almost 4,000 with diabetes. Overall, they found that aspirin reduced the risk of serious vascular events by 12% (relative risk 0.88 [95% CI 0.82–0.94]). The largest reduction was for nonfatal MI, with little effect on CHD death (relative risk 0.95 [95% CI 0.78–1.15]) or total stroke.

Most recently, the ASCEND (A Study of Cardiovascular Events iN Diabetes) trial randomized 15,480 people with diabetes but no evident cardiovascular disease to aspirin 100 mg daily or placebo (144). The primary efficacy end point was vascular death, MI, or stroke or transient ischemic attack. The primary safety outcome was major bleeding (i.e., intracranial hemorrhage, sight-threatening bleeding in the eye, gastrointestinal bleeding, or other serious bleeding). During a mean follow-up of 7.4 years, there was a significant 12% reduction in the primary efficacy end point (8.5% vs. 9.6%; P = 0.01). In contrast, major bleeding was significantly increased from 3.2 to 4.1% in the aspirin group (rate ratio 1.29; P = 0.003), with most of the excess being gastrointestinal bleeding and other extracranial bleeding. There were no significant differences by sex, weight, or duration of diabetes or other baseline factors including ASCVD risk score.

Two other large, randomized trials of aspirin for primary prevention, in people without diabetes (ARRIVE [Aspirin to Reduce Risk of Initial Vascular Events]) (145) and in the elderly (ASPREE [Aspirin in Reducing Events in the Elderly]) (146), which included 11% with diabetes, found no benefit of aspirin on the primary efficacy end point and an increased risk of bleeding. In ARRIVE, with 12,546 patients over a period of 60 months follow-up, the primary end point occurred in 4.29% vs. 4.48% of patients in the aspirin versus placebo groups (HR 0.96 [95% CI 0.81–1.13]; P = 0.60). Gastrointestinal bleeding events (characterized as mild) occurred in 0.97% of patients in the aspirin group vs. 0.46% in the placebo group (HR 2.11 [95% CI 1.36–3.28]; P = 0.0007). In ASPREE, including 19,114 individuals, for cardiovascular disease (fatal CHD, MI, stroke, or hospitalization for heart failure) after a median of 4.7 years of follow-up, the rates per 1,000 person-years were 10.7 vs. 11.3 events in aspirin vs. placebo groups (HR 0.95 [95% CI 0.83–1.08]). The rate of major hemorrhage per 1,000 person-years was 8.6 events vs. 6.2 events, respectively (HR 1.38 [95% CI 1.18–1.62]; P < 0.001).

Thus, aspirin appears to have a modest effect on ischemic vascular events, with the absolute decrease in events depending on the underlying ASCVD risk. The main adverse effect is an increased risk of gastrointestinal bleeding. The excess risk may be as high as 5 per 1,000 per year in real-world settings. However, for adults with ASCVD risk >1% per year, the number of ASCVD events prevented will be similar to the number of episodes of bleeding induced, although these complications do not have equal effects on long-term health (147).

Recommendations for using aspirin as primary prevention include both men and women aged ≥50 years with diabetes and at least one additional major risk factor (family history of premature ASCVD, hypertension, dyslipidemia, smoking, or CKD/albuminuria) who are not at increased risk of bleeding (e.g., older age, anemia, renal disease) (148151). Noninvasive imaging techniques such as coronary calcium scoring may potentially help further tailor aspirin therapy, particularly in those at low risk (152,153). For people >70 years of age (with or without diabetes), the balance appears to have greater risk than benefit (144,146). Thus, for primary prevention, the use of aspirin needs to be carefully considered and may generally not be recommended. Aspirin may be considered in the context of high cardiovascular risk with low bleeding risk, but generally not in older adults. Aspirin therapy for primary prevention may be considered in the context of shared decision-making, which carefully weighs the cardiovascular benefits with the fairly comparable increase in risk of bleeding.

For people with documented ASCVD, use of aspirin for secondary prevention has far greater benefit than risk; for this indication, aspirin is still recommended (139).

Aspirin Use in People <50 Years of Age

Aspirin is not recommended for those at low risk of ASCVD (such as men and women aged <50 years with diabetes with no other major ASCVD risk factors) as the low benefit is likely to be outweighed by the risks of bleeding. Clinical judgment should be used for those at intermediate risk (younger patients with one or more risk factors or older patients with no risk factors) until further research is available. Patients’ willingness to undergo long-term aspirin therapy should also be considered (154). Aspirin use in patients aged <21 years is generally contraindicated due to the associated risk of Reye syndrome.

Aspirin Dosing

Average daily dosages used in most clinical trials involving people with diabetes ranged from 50 mg to 650 mg but were mostly in the range of 100–325 mg/day. There is little evidence to support any specific dose but using the lowest possible dose may help to reduce side effects (155). In the ADAPTABLE (Aspirin Dosing: A Patient-Centric Trial Assessing Benefits and Long-term Effectiveness) trial of individuals with established cardiovascular disease, 38% of whom had diabetes, there were no significant differences in cardiovascular events or major bleeding between patients assigned to 81 mg and those assigned to 325 mg of aspirin daily (156). In the U.S., the most common low-dose tablet is 81 mg. Although platelets from people with diabetes have altered function, it is unclear what, if any, effect that finding has on the required dose of aspirin for cardioprotective effects in people with diabetes. Many alternate pathways for platelet activation exist that are independent of thromboxane A2 and thus are not sensitive to the effects of aspirin (157). “Aspirin resistance” has been described in people with diabetes when measured by a variety of ex vivo and in vitro methods (platelet aggregometry, measurement of thromboxane B2) (158), but other studies suggest no impairment in aspirin response among people with diabetes (159). A trial suggested that more frequent dosing regimens of aspirin may reduce platelet reactivity in individuals with diabetes (160); however, these observations alone are insufficient to empirically recommend that higher doses of aspirin be used in this group at this time. Another meta-analysis raised the hypothesis that low-dose aspirin efficacy is reduced in those weighing >70 kg (161); however, the ASCEND trial found benefit of low-dose aspirin in those in this weight range, which would thus not validate this suggested hypothesis (144). It appears that 75–162 mg/day is optimal.

Indications for P2Y12 Receptor Antagonist Use

A P2Y12 receptor antagonist in combination with aspirin is reasonable for at least 1 year in patients following an ACS and may have benefits beyond this period. Evidence supports use of either ticagrelor or clopidogrel if no percutaneous coronary intervention was performed and clopidogrel, ticagrelor, or prasugrel if a percutaneous coronary intervention was performed (162). In people with diabetes and prior MI (1–3 years before), adding ticagrelor to aspirin significantly reduces the risk of recurrent ischemic events including cardiovascular and CHD death (163). Similarly, the addition of ticagrelor to aspirin reduced the risk of ischemic cardiovascular events compared with aspirin alone in people with diabetes and stable coronary artery disease (164,165). However, a higher incidence of major bleeding, including intracranial hemorrhage, was noted with dual antiplatelet therapy. The net clinical benefit (ischemic benefit vs. bleeding risk) was improved with ticagrelor therapy in the large prespecified subgroup of patients with history of percutaneous coronary intervention, while no net benefit was seen in patients without prior percutaneous coronary intervention (165). However, early aspirin discontinuation compared with continued dual antiplatelet therapy after coronary stenting may reduce the risk of bleeding without a corresponding increase in the risks of mortality and ischemic events, as shown in a prespecified analysis of people with diabetes enrolled in the TWILIGHT (Ticagrelor With Aspirin or Alone in High-Risk Patients After Coronary Intervention) trial and a recent meta-analysis (166,167).

Combination Antiplatelet and Anticoagulation Therapy

Combination therapy with aspirin plus low dose rivaroxaban may be considered for people with stable coronary and/or peripheral artery disease to prevent major adverse limb and cardiovascular complications. In the COMPASS (Cardiovascular Outcomes for People Using Anticoagulation Strategies) trial of 27,395 individuals with established coronary artery disease and/or peripheral artery disease, aspirin plus rivaroxaban 2.5 mg twice daily was superior to aspirin plus placebo in the reduction of cardiovascular ischemic events including major adverse limb events. The absolute benefits of combination therapy appeared larger in people with diabetes, who comprised 10,341 of the trial participants (168,169). A similar treatment strategy was evaluated in the Vascular Outcomes Study of ASA (acetylsalicylic acid) Along with Rivaroxaban in Endovascular or Surgical Limb Revascularization for Peripheral Artery Disease (VOYAGER PAD) trial (170), in which 6,564 individuals with peripheral artery disease who had undergone revascularization were randomly assigned to receive rivaroxaban 2.5 mg twice daily plus aspirin or placebo plus aspirin. Rivaroxaban treatment in this group of patients was also associated with a significantly lower incidence of ischemic cardiovascular events, including major adverse limb events. However, an increased risk of major bleeding was noted with rivaroxaban added to aspirin treatment in both COMPASS and VOYAGER PAD.

The risks and benefits of dual antiplatelet or antiplatelet plus anticoagulant treatment strategies should be thoroughly discussed with eligible patients, and shared decision-making should be used to determine an individually appropriate treatment approach. This field of cardiovascular risk reduction is evolving rapidly, as are the definitions of optimal care for patients with differing types and circumstances of cardiovascular complications.

Cardiovascular Disease

Screening

Recommendations

  • 10.39 In asymptomatic individuals, routine screening for coronary artery disease is not recommended as it does not improve outcomes as long as atherosclerotic cardiovascular disease risk factors are treated. A

  • 10.40 Consider investigations for coronary artery disease in the presence of any of the following: atypical cardiac symptoms (e.g., unexplained dyspnea, chest discomfort); signs or symptoms of associated vascular disease including carotid bruits, transient ischemic attack, stroke, claudication, or peripheral arterial disease; or electrocardiogram abnormalities (e.g., Q waves). E

Treatment

Recommendations

  • 10.41 Among people with type 2 diabetes who have established atherosclerotic cardiovascular disease or established kidney disease, a sodium–glucose cotransporter 2 inhibitor or glucagon-like peptide 1 receptor agonist with demonstrated cardiovascular disease benefit (Table 10.3B and Table 10.3C) is recommended as part of the comprehensive cardiovascular risk reduction and/or glucose-lowering regimens. A

  • 10.41a In people with type 2 diabetes and established atherosclerotic cardiovascular disease, multiple atherosclerotic cardiovascular disease risk factors, or diabetic kidney disease, a sodium–glucose cotransporter 2 inhibitor with demonstrated cardiovascular benefit is recommended to reduce the risk of major adverse cardiovascular events and/or heart failure hospitalization. A

  • 10.41b In people with type 2 diabetes and established atherosclerotic cardiovascular disease or multiple risk factors for atherosclerotic cardiovascular disease, a glucagon-like peptide 1 receptor agonist with demonstrated cardiovascular benefit is recommended to reduce the risk of major adverse cardiovascular events. A

  • 10.41c In people with type 2 diabetes and established atherosclerotic cardiovascular disease or multiple risk factors for atherosclerotic cardiovascular disease, combined therapy with a sodium–glucose cotransporter 2 inhibitor with demonstrated cardiovascular benefit and a glucagon-like peptide 1 receptor agonist with demonstrated cardiovascular benefit may be considered for additive reduction in the risk of adverse cardiovascular and kidney events. A

  • 10.42a In people with type 2 diabetes and established heart failure with either preserved or reduced ejection fraction, a sodium–glucose cotransporter 2 inhibitor with proven benefit in this patient population is recommended to reduce risk of worsening heart failure and cardiovascular death. A

  • 10.42b In people with type 2 diabetes and established heart failure with either preserved or reduced ejection fraction, a sodium–glucose cotransporter 2 inhibitor with proven benefit in this patient population is recommended to improve symptoms, physical limitations, and quality of life. A

  • 10.43 For people with type 2 diabetes and chronic kidney disease with albuminuria treated with maximum tolerated doses of ACE inhibitor or angiotensin receptor blocker, addition of finerenone is recommended to improve cardiovascular outcomes and reduce the risk of chronic kidney disease progression. A

  • 10.44 In people with known atherosclerotic cardiovascular disease, particularly coronary artery disease, ACE inhibitor or angiotensin receptor blocker therapy is recommended to reduce the risk of cardiovascular events. A

  • 10.45 In people with prior myocardial infarction, β-blockers should be continued for 3 years after the event. B

  • 10.46 Treatment of individuals with heart failure with reduced ejection fraction should include a β-blocker with proven cardiovascular outcomes benefit, unless otherwise contraindicated. A

  • 10.47 In people with type 2 diabetes with stable heart failure, metformin may be continued for glucose lowering if estimated glomerular filtration rate remains >30 mL/min/1.73 m2 but should be avoided in unstable or hospitalized individuals with heart failure. B

Table 10.3B.

Cardiovascular and cardiorenal outcomes trials of available antihyperglycemic medications completed after the issuance of the FDA 2008 guidelines: GLP-1 receptor agonists

ELIXA (208) LEADER (203) SUSTAIN-6 (204)* EXSCEL (209) REWIND (207) PIONEER-6 (205)
(n = 6,068) (n = 9,340) (n = 3,297) (n = 14,752) (n = 9,901) (n = 3,183)
Intervention Lixisenatide/placebo Liraglutide/placebo Semaglutide s.c. injection/placebo Exenatide QW/ placebo Dulaglutide/placebo Semaglutide oral/ placebo
Main inclusion criteria Type 2 diabetes and history of ACS (<180 days) Type 2 diabetes and preexisting CVD, CKD, or HF at ≥50 years of age or CV risk at ≥60 years of age Type 2 diabetes and preexisting CVD, HF, or CKD at ≥50 years of age or CV risk at ≥60 years of age Type 2 diabetes with or without preexisting CVD Type 2 diabetes and prior ASCVD event or risk factors for ASCVD Type 2 diabetes and high CV risk (age of ≥50 years with established CVD or CKD, or age of ≥60 years with CV risk factors only)
A1C inclusion criteria (%) 5.5–11.0 ≥7.0 ≥7.0 6.5–10.0 ≤9.5 None
Age (years) 60.3 64.3 64.6 62 66.2 66
Race (% White) 75.2 77.5 83.0 75.8 75.7 72.3
Sex (% male) 69.3 64.3 60.7 62 53.7 68.4
Diabetes duration (years) 9.3 12.8 13.9 12 10.5 14.9
Median follow-up (years) 2.1 3.8 2.1 3.2 5.4 1.3
Statin use (%) 93 72 73 74 66 85.2 (all lipid-lowering)
Metformin use (%) 66 76 73 77 81 77.4
Prior CVD/CHF (%) 100/22 81/18 60/24 73.1/16.2 32/9 84.7/12.2
Mean baseline A1C (%) 7.7 8.7 8.7 8.0 7.4 8.2
Mean difference in A1C between groups at end of treatment (%) −0.3^ −0.4 −0.7 or −1.0^ −0.53^ −0.61 −0.7
Year started/reported 2010/2015 2010/2016 2013/2016 2010/2017 2011/2019 2017/2019
Primary outcome§ 4-point MACE 1.02 (0.89–1.17) 3-point MACE 0.87 (0.78–0.97) 3-point MACE 0.74 (0.58–0.95) 3-point MACE 0.91 (0.83–1.00) 3-point MACE 0.88 (0.79–0.99) 3-point MACE 0.79 (0.57–1.11)
Key secondary outcome§ Expanded MACE 1.02 (0.90–1.11) Expanded MACE 0.88 (0.81–0.96) Expanded MACE 0.74 (0.62–0.89) Individual components of MACE (see below) Composite microvascular outcome (eye or renal outcome) 0.87 (0.79–0.95) Expanded MACE or HF hospitalization 0.82 (0.61–1.10)
Cardiovascular death§ 0.98 (0.78–1.22) 0.78 (0.66–0.93) 0.98 (0.65–1.48) 0.88 (0.76–1.02) 0.91 (0.78–1.06) 0.49 (0.27–0.92)
Ml§ 1.03 (0.87–1.22) 0.86 (0.73–1.00) 0.74 (0.51–1.08) 0.97 (0.85–1.10) 0.96 (0.79–1.15) 1.18 (0.73–1.90)
Stroke§ 1.12 (0.79–1.58) 0.86 (0.71–1.06) 0.61 (0.38–0.99) 0.85 (0.70–1.03) 0.76 (0.61–0.95) 0.74 (0.35–1.57)
HF hospitalization§ 0.96 (0.75–1.23) 0.87 (0.73–1.05) 1.11 (0.77–1.61) 0.94 (0.78–1.13) 0.93 (0.77–1.12) 0.86 (0.48–1.55)
Unstable angina hospitalization§ 1.11 (0.47–2.62) 0.98 (0.76–1.26) 0.82 (0.47–1.44) 1.05 (0.94–1.18) 1.14 (0.84–1.54) 1.56 (0.60–4.01)
All-cause mortality§ 0.94 (0.78–1.13) 0.85 (0.74–0.97) 1.05 (0.74–1.50) 0.86 (0.77–0.97) 0.90 (0.80–1.01) 0.51 (0.31–0.84)
Worsening nephropathy§ǁ 0.78 (0.67–0.92) 0.64 (0.46–0.88) 0.85 (0.77–0.93)

—, not assessed/reported; ACS, acute coronary syndrome; ASCVD, atherosclerotic cardiovascular disease; CHF, congestive heart failure; CKD, chronic kidney disease; CV, cardiovascular; CVD, cardiovascular disease; GLP-1, glucagon-like peptide 1; HF, heart failure; MACE, major adverse cardiovascular event; Ml, myocardial infarction. Data from this table was adapted from Cefalu et al. (238) in the January 2018 issue of Diabetes Care.

*

Powered to rule out a hazard ratio of 1.8; superiority hypothesis not prespecified.

Age was reported as means in all trials; diabetes duration was reported as means in all trials except EXSCEL, which reported medians.

Significant difference in A1C between groups (P < 0.05).

^

AIC change of 0.66% with 0.5 mg and 1.05% with 1 mg dose of semaglutide.

§

Outcomes reported as hazard ratio (95% Cl).

ǁ

Worsening nephropathy is defined as the new onset of urine albumin-to-creatinine ratio >300 mg/g creatinine or a doubling of the serum creatinine level and an estimated glomerular filtration rate of <45 mL/min/1.73 m2, the need for continuous renal replacement therapy, or death from renal disease in LEADER and SUSTAIN-6 and as new macroalbuminuria, a sustained decline in estimated glomerular filtration rate of 30% or more from baseline, or chronic renal replacement therapy in REWIND. Worsening nephropathy was a prespecified exploratory adjudicated outcome in LEADER, SUSTAIN-6, and REWIND.

Table 10.3C.

Cardiovascular and cardiorenal outcomes trials of available antihyperglycemic medications completed after the issuance of the FDA 2008 guidelines: SGLT2 inhibitors

EMPA-REG OUTCOME (8) (n = 7,020) CANVAS Program (9) (n = 10,142) DECLARE-TIMI 58 (196) (n = 17,160) CREDENCE (194) (n = 4,401) DAPA-CKD (197,239) (n = 4,304; 2,906 with diabetes) VERTIS CV (201,240) (n = 8,246) DAPA-HF (11) (n = 4,744; 1,983 with diabetes) EMPEROR-Reduced (200) (n = 3,730; 1,856 with diabetes) EMPEROR-Preserved (189,241) (n = 5,988; 2,938 with diabetes) DELIVER (199) (n = 6,263; 2,807 with diabetes)
Intervention Empagliflozin/placebo Canagliflozin/placebo Dapagliflozin/placebo Canagliflozin/placebo Dapagliflozin/placebo Ertugliflozin/placebo Dapagliflozin/placebo Empagliflozin/placebo* Empagliflozin/placebo Dapagliflozin/placebo
Main inclusion criteria Type 2 diabetes and preexisting CVD Type 2 diabetes and preexisting CVD at ≥30 years of age or ≥2 CV risk factors at ≥50 years of age Type 2 diabetes and established ASCVD or multiple risk factors for ASCVD Type 2 diabetes and albuminuric kidney disease Albuminuric kidney disease, with or without diabetes Type 2 diabetes and ASCVD NYHA class II, III, or IV heart failure and an ejection fraction ≤40%, with or without diabetes NYHA class II, III, or IV heart failure and an ejection fraction ≤40%, with or without diabetes NYHA class II, III, or IV heart failure and an ejection fraction >40% NYHA class II, III, or IV heart failure and an ejection fraction >40% with or without diabetes
A1C inclusion criteria (%) 7.0–10.0 7.0–10.5 ≥6.5 6.5–12 7.0–10.5
Age (years) 63.1 63.3 64.0 63 61.8 64.4 66 67.2, 66.5 71.8, 71.9 71.7
Race (% White) 72.4 78.3 79.6 66.6 53.2 87.8 70.3 71.1, 69.8 76.3, 75.4 71.2
Sex (% male) 71.5 64.2 62.6 66.1 66.9 70 76.6 76.5, 75.6 55.4, 55.3 56.1
Diabetes duration (years) 57% >10 13.5 11.0 15.8 12.9
Median follow-up (years) 3.1 3.6 4.2 2.6 2.4 3.5 1.5 1.3 2.2 2.3
Statin use (%) 77 75 75 (statin or ezetimibe use) 69 64.9 68.1, 68.8
Metformin use (%) 74 77 82 57.8 29 51.2% (of people with diabetes)
Prior CVD/CHF (%) 99/10 65.6/14.4 40/10 50.4/14.8 37.4/10.9 99.9/23.1 100% with CHF 100% with CHF 100% with CHF 100% with CHF
Mean baseline A1C (%) 8.1 8.2 8.3 8.3 7.1% (7.8% in those with diabetes) 8.2 6.6
Mean difference in A1C between groups at end of treatment (%) −0.3^ −0.58 −0.43 −0.31 −0.48 to −0.5
Year started/reported 2010/2015 2009/2017 2013/2018 2017/2019 2017/2020 2013/2020 2017/2019 2017/2020 2017/2020 2018/2022
Primary outcome§ 3-point MACE 0.86 (0.74–0.99) 3-point MACE 0.86 (0.75–0.97) 3-point MACE 0.93 (0.84–1.03)
CV death or HF hospitalization 0.83 (0.73–0.95)
ESRD, doubling of creatinine, or death from renal or CV cause 0.70 (0.59–0.82) ≥50% decline in eGFR, ESKD, or death from renal or CV cause 0.61 (0.51–0.72) 3-point MACE 0.97 (0.85–1.11) Worsening heart failure or death from CV causes 0.74 (0.65–0.85)
Results did not differ by diabetes status
CV death or HF hospitalization 0.75 (0.65–0.86) CV death or HF hospitalization 0.79 (0.69–0.90) Worsening HF or CV death 0.82 (0.73–0.92)
Key secondary outcome§ 4-point MACE 0.89 (0.78–1.01) All-cause and CV mortality (see below) Death from any cause 0.93 (0.82–1.04)
Renal composite (≥40% decrease in eGFR rate to <60 mL/min/1.73 m2, new ESRD, or death from renal or CV causes 0.76 (0.67–0.87)
CV death or HF hospitalization 0.69 (0.57–0.83)
3-point MACE 0.80 (0.67–0.95)
≥50% decline in eGFR, ESKD, or death from renal cause 0.56 (0.45–0.68)
CV death or HF hospitalization 0.71 (0.55–0.92)
Death from any cause 0.69 (0.53–0.88)
CV death or HF hospitalization 0.88 (0.75–1.03)
CV death 0.92 (0.77–1.11)
Renal death, renal replacement therapy, or doubling of creatinine 0.81 (0.63–1.04)
CV death or HF hospitalization 0.75 (0.65–0.85) Total HF hospitalizations 0.70 (0.58–0.85)
Mean slope of change in eGFR 1.73 (1.10–2.37)
All HF hospitalizations (first and recurrent) 0.73 (0.61–0.88)
Rate of decline in eGFR (−1.25 vs. −2.62 mL/min/1.73 m2; P < 0.001)
Total number worsening HF and CV deaths 0.77 (0.67–0.89)
Change in KCCQ TSS at month 8 1.11 (1.03–1.21)
Mean change in KCCQ TSS 2.4 (1.5–3.4)
All-cause mortality 0.94 (0.83–1.07)
Cardiovascular death§ 0.62 (0.49–0.77) 0.87 (0.72–1.06) 0.98 (0.82–1.17) 0.78 (0.61–1.00) 0.81 (0.58–1.12) 0.92 (0.77–1.11) 0.82 (0.69–0.98) 0.92 (0.75–1.12) 0.91 (0.76–1.09) 0.88 (0.74–1.05)
MI§ 0.87 (0.70–1.09) 0.89 (0.73–1.09) 0.89 (0.77–1.01) 1.04 (0.86–1.26)
Stroke§ 1.18 (0.89–1.56) 87 (0.69–1.09) 1.01 (0.84–1.21) 1.06 (0.82–1.37)
HF hospitalization§ 0.65 (0.50–0.85) 67 (0.52–0.87) 0.73 (0.61–0.88) 0.61 (0.47–0.80) 0.70 (0.54–0.90) 0.70 (0.59–0.83) 0.69 (0.59–0.81) 0.73 (0.61–0.88) 0.77 (0.67–0.89)
Unstable angina hospitalization§ 0.99 (0.74–1.34)
All-cause mortality§ 0.68 (0.57–0.82) 87 (0.74–1.01) 0.93 (0.82–1.04) 0.83 (0.68–1.02) 0.69 (0.53–0.88) 0.93 (0.80–1.08) 0.83 (0.71–0.97) 0.92 (0.77–1.10) 1.00 (0.87–1.15) 0.94 (0.83–1.07)
Worsening nephropathy§ǁ 0.61 (0.53–0.70) 0.60 (0.47–0.77) 0.53 (0.43–0.66) (See primary outcome) (See primary outcome) (See secondary outcomes) 0.71 (0.44–1.16) Composite renal outcome 0.50 (0.32–0.77) Composite renal outcome** 0.95 (0.73–1.24)

—, not assessed/reported; CHF, congestive heart failure; CV, cardiovascular; CVD, cardiovascular disease; eGFR, estimated glomerular filtration rate; ESRD, end-stage renal disease; HF, heart failure; KCCQ TSS, Kansas City Cardiomyopathy Questionnaire Total Symptom Score; MACE, major adverse cardiovascular event; Ml, myocardial infarction; SGLT2, sodium–glucose cotransporter 2; NYFIA, New York Fleart Association. Data from this table was adapted from Cefalu et al. (238) in the January 2018 issue of Diabetes Care.

*

Baseline characteristics for EMPEROR-Reduced displayed as empagliflozin, placebo.

Age was reported as means in all trials; diabetes duration was reported as means in all trials except EMPA-REG OUTCOME, which reported as percentage of population with diabetes duration >10 years, and DECLARE-TIMI 58, which reported median.

Significant difference in A1C between groups (P < 0.05).

^

AIC change of 0.30 in EMPA-REG OUTCOME is based on pooled results for both doses (i.e., 0.24% for 10 mg and 0.36% for 25 mg of empagliflozin).

§

Outcomes reported as hazard ratio (95% Cl).

ǁ

Definitions of worsening nephropathy differed between trials.

**

Composite outcome in EMPEROR-Preserved: time to first occurrence of chronic dialysis, renal transplantation; sustained reduction of ≥40% in eGFR, sustained eGFR <15 mL/min/1.73 m2 for individuals with baseline eGFR ≥30 mL/min/1.73 m2.

Cardiac Testing

Candidates for advanced or invasive cardiac testing include those with 1) typical or atypical cardiac symptoms and 2) an abnormal resting electrocardiogram (ECG). Exercise ECG testing without or with echocardiography may be used as the initial test. In adults with diabetes ≥40 years of age, measurement of coronary artery calcium is also reasonable for cardiovascular risk assessment. Pharmacologic stress echocardiography or nuclear imaging should be considered in individuals with diabetes in whom resting ECG abnormalities preclude exercise stress testing (e.g., left bundle branch block or ST-T abnormalities). In addition, individuals who require stress testing and are unable to exercise should undergo pharmacologic stress echocardiography or nuclear imaging.

Screening Asymptomatic Patients

The screening of asymptomatic patients with high ASCVD risk is not recommended (171), in part because these high-risk patients should already be receiving intensive medical therapy—an approach that provides benefit similar to invasive revascularization (172,173). There is also some evidence that silent ischemia may reverse over time, adding to the controversy concerning aggressive screening strategies (174). In prospective studies, coronary artery calcium has been established as an independent predictor of future ASCVD events in people with diabetes and is consistently superior to both the UK Prospective Diabetes Study (UKPDS) risk engine and the Framingham Risk Score in predicting risk in this population (175177). However, a randomized observational trial demonstrated no clinical benefit to routine screening of asymptomatic people with type 2 diabetes and normal ECGs (178). Despite abnormal myocardial perfusion imaging in more than one in five patients, cardiac outcomes were essentially equal (and very low) in screened versus unscreened patients. Accordingly, indiscriminate screening is not considered cost-effective. Studies have found that a risk factor-based approach to the initial diagnostic evaluation and subsequent follow-up for coronary artery disease fails to identify which people with type 2 diabetes will have silent ischemia on screening tests (179,180).

Any benefit of newer noninvasive coronary artery disease screening methods, such as computed tomography calcium scoring and computed tomography angiography, to identify patient subgroups for different treatment strategies remains unproven in asymptomatic people with diabetes, though research is ongoing. Since asymptomatic people with diabetes with higher coronary disease burden have more future cardiac events (175,181,182), these additional imaging tests may provide reasoning for treatment intensification and/or guide informed patient decision-making and willingness for medication initiation and participation.

While coronary artery screening methods, such as calcium scoring, may improve cardiovascular risk assessment in people with type 2 diabetes (183), their routine use leads to radiation exposure and may result in unnecessary invasive testing such as coronary angiography and revascularization procedures. The ultimate balance of benefit, cost, and risks of such an approach in asymptomatic patients remains controversial, particularly in the modern setting of aggressive ASCVD risk factor control.

Lifestyle and Pharmacologic Interventions

Intensive lifestyle intervention focusing on weight loss through decreased caloric intake and increased physical activity as performed in the Action for Health in Diabetes (Look AHEAD) trial may be considered for improving glucose control, fitness, and some ASCVD risk factors (184). Patients at increased ASCVD risk should receive statin, ACE inhibitor, or ARB therapy if the patient has hypertension, and possibly aspirin, unless there are contraindications to a particular drug class. Clear benefit exists for ACE inhibitor or ARB therapy in people with diabetic kidney disease or hypertension, and these agents are recommended for hypertension management in people with known ASCVD (particularly coronary artery disease) (65,66,185). People with type 2 diabetes and CKD should be considered for treatment with finerenone to reduce cardiovascular outcomes and the risk of CKD progression (186189). β-Blockers should be used in individuals with active angina or HFrEF and for 3 years after Ml in those with preserved left ventricular function (190,191).

Glucose-Lowering Therapies and Cardiovascular Outcomes

In 2008, the FDA issued a guidance for industry to perform cardiovascular outcomes trials for all new medications for the treatment for type 2 diabetes amid concerns of increased cardiovascular risk (192). Previously approved diabetes medications were not subject to the guidance. Recently published cardiovascular outcomes trials have provided additional data on cardiovascular and renal outcomes in people with type 2 diabetes with cardiovascular disease or at high risk for cardiovascular disease (Table 10.3A, Table 10.3B, and Table 10.3C). An expanded review of the effects of glucose-lowering and other therapies in people with CKD is included in Section 11, “Chronic Kidney Disease and Risk Management.”

Table 10.3A.

Cardiovascular and cardiorenal outcomes trials of available antihyperglycemic medications completed after the issuance of the FDA 2008 guidelines: DPP-4 inhibitors

SAVOR-TIMI 53 (224) EXAMINE (235) TECOS (226) CARMELINA (193,236) CAROLINA (193,237)
(n = 16,492) (n = 5,380) (n = 14,671) (n = 6,979) (n = 6,042)
Intervention Saxagliptin/placebo Alogliptin/placebo Sitagliptin/placebo Linagliptin/placebo Linagliptin/glimepiride
Main inclusion criteria Type 2 diabetes and history of or multiple risk factors for CVD Type 2 diabetes and ACS within 15–90 days before randomization Type 2 diabetes and preexisting CVD Type 2 diabetes and high CV and renal risk Type 2 diabetes and high CV risk
A1C inclusion criteria (%) ≥6.5 6.5–11.0 6.5–8.0 6.5–10.0 6.5–8.5
Age (years) 65.1 61.0 65.4 65.8 64.0
Race (% White) 75.2 72.7 67.9 80.2 73.0
Sex (% male) 66.9 67.9 70.7 62.9 60.0
Diabetes duration (years) 10.3 7.1 11.6 14.7 6.2
Median follow-up (years) 2.1 1.5 3.0 2.2 6.3
Statin use (%) 78 91 80 71.8 64.1
Metformin use (%) 70 66 82 54.8 82.5
Prior CVD/CHF (%) 78/13 100/28 74/18 57/26.8 34.5/4.5
Mean baseline A1C (%) 8.0 8.0 7.2 7.9 7.2
Mean difference in A1C between groups at end of treatment (%) −0.3 −0.3 −0.3 −0.36 0
Year started/reported 2010/2013 2009/2013 2008/2015 2013/2018 2010/2019
Primary outcome§ 3-point MACE 1.00 (0.89–1.12) 3-point MACE 0.96 (95% UL ≤1.16) 4-point MACE 0.98 (0.89–1.08) 3-point MACE 1.02 (0.89–1.17) 3-point MACE 0.98 (0.84–1.14)
Key secondary outcome§ Expanded MACE 1.02 (0.94–1.11) 4-point MACE 0.95 (95% UL ≤1.14) 3-point MACE 0.99 (0.89–1.10) Kidney composite (ESRD, sustained ≥40% decrease in eGFR, or renal death) 1.04 (0.89–1.22) 4-point MACE 0.99 (0.86–1.14)
Cardiovascular death§ 1.03 (0.87–1.22) 0.85 (0.66–1.10) 1.03 (0.89–1.19) 0.96 (0.81–1.14) 1.00 (0.81–1.24)
MI§ 0.95 (0.80–1.12) 1.08 (0.88–1.33) 0.95 (0.81–1.11) 1.12 (0.90–1.40) 1.03 (0.82–1.29)
Stroke§ 1.11 (0.88–1.39) 0.91 (0.55–1.50) 0.97 (0.79–1.19) 0.91 (0.67–1.23) 0.86 (0.66–1.12)
HF hospitalization§ 1.27 (1.07–1.51) 1.19 (0.90–1.58) 1.00 (0.83–1.20) 0.90 (0.74–1.08) 1.21 (0.92–1.59)
Unstable angina hospitalization§ 1.19 (0.89–1.60) 0.90 (0.60–1.37) 0.90 (0.70–1.16) 0.87 (0.57–1.31) 1.07 (0.74–1.54)
All-cause mortality§ 1.11 (0.96–1.27) 0.88 (0.71–1.09) 1.01 (0.90–1.14) 0.98 (0.84–1.13) 0.91 (0.78–1.06)
Worsening nephropathy§ǁ 1.08 (0.88–1.32) Kidney composite (see above)

—, not assessed/reported; ACS, acute coronary syndrome; CHF, congestive heart failure; CV, cardiovascular; CVD, cardiovascular disease; DPP-4, dipeptidyl peptidase 4; eGFR, estimated glomerular filtration rate; ESRD, end-stage renal disease; GLP-1, glucagon-like peptide 1; HF, heart failure; MACE, major adverse cardiovascular event; MI, myocardial infarction; UL, upper limit. Data from this table was adapted from Cefalu et al. (238) in the January 2018 issue of Diabetes Care.

Age was reported as means in all trials except EXAMINE, which reported medians; diabetes duration was reported as means in all trials except SAVOR-TIMI 53 and EXAMINE, which reported medians.

Significant difference in A1C between groups (P < 0.05).

§

Outcomes reported as hazard ratio (95% CI).

ǁ

Worsening nephropathy is defined as a doubling of creatinine level, initiation of dialysis, renal transplantation, or creatinine >6.0 mg/dL (530 mmol/L) in SAVOR-TIMI 53. Worsening nephropathy was a prespecified exploratory adjudicated outcome in SAVOR-TIMI 53.

Cardiovascular outcomes trials of dipeptidyl peptidase 4 (DPP-4) inhibitors have all, so far, not shown cardiovascular benefits relative to placebo. In addition, the CAROLINA (Cardiovascular Outcome Study of Linagliptin Versus Glimepiride in Type 2 Diabetes) study demonstrated noninferiority between a DPP-4 inhibitor, linagliptin, and a sulfonylurea, glimepiride, on cardiovascular outcomes despite lower rates of hypoglycemia in the linagliptin treatment group (193). However, results from other new agents have provided a mix of results.

SGLT2 Inhibitor Trials

The Bl 10773 (Empagliflozin) Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients (EMPA-REG OUTCOME) was a randomized, double-blind trial that assessed the effect of empagliflozin, an SGLT2 inhibitor, versus placebo on cardiovascular outcomes in 7,020 people with type 2 diabetes and existing cardiovascular disease. Study participants had a mean age of 63 years, 57% had diabetes for more than 10 years, and 99% had established cardiovascular disease. EMPA-REG OUTCOME showed that over a median follow-up of 3.1 years, treatment reduced the composite outcome of MI, stroke, and cardiovascular death by 14% (absolute rate 10.5% vs. 12.1% in the placebo group, HR in the empagliflozin group 0.86 [95% CI 0.74–0.99]; P = 0.04 for superiority) and cardiovascular death by 38% (absolute rate 3.7% vs. 5.9%, HR 0.62 [95% CI 0.49–0.77]; P < 0.001) (8).

Two large outcomes trials of the SGLT2 inhibitor canagliflozin have been conducted that separately assessed 1) the cardiovascular effects of treatment in patients at high risk for major adverse cardiovascular events (9) and 2) the impact of canagliflozin therapy on cardiorenal outcomes in people with diabetes-related CKD (194). First, the Canagliflozin Cardiovascular Assessment Study (CANVAS) Program integrated data from two trials. The CANVAS trial that started in 2009 was partially unblinded prior to completion because of the need to file interim cardiovascular outcomes data for regulatory approval of the drug (195). Thereafter, the post approval CANVAS-Renal (CANVAS-R) trial was started in 2014. Combining both trials, 10,142 participants with type 2 diabetes were randomized to canagliflozin or placebo and were followed for an average 3.6 years. The mean age of patients was 63 years, and 66% had a history of cardiovascular disease. The combined analysis of the two trials found that canagliflozin significantly reduced the composite outcome of cardiovascular death, MI, or stroke versus placebo (occurring in 26.9 vs. 31.5 participants per 1,000 patient-years; HR 0.86 [95% CI 0.75–0.97]). The specific estimates for canagliflozin versus placebo on the primary composite cardiovascular outcome were HR 0.88 (95% CI 0.75–1.03) for the CANVAS trial and 0.82 (0.66–1.01) for CANVAS-R, with no heterogeneity found between trials. Of note, there was an increased risk of lower-limb amputation with canagliflozin (6.3 vs. 3.4 participants per 1,000 patient-years; HR 1.97 [95% CI 1.41–2.75]) (9). Second, the Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation (CREDENCE) trial randomized 4,401 people with type 2 diabetes and chronic diabetes-related kidney disease (UACR >300 mg/g and eGFR 30 to <90 mL/min/1.73 m2) to canagliflozin 100 mg daily or placebo (194). The primary outcome was a composite of end-stage kidney disease, doubling of serum creatinine, or death from renal or cardiovascular causes. The trial was stopped early due to conclusive evidence of efficacy identified during a prespecified interim analysis with no unexpected safety signals. The risk of the primary composite outcome was 30% lower with canagliflozin treatment when compared with placebo (HR 0.70 [95% CI 0.59–0.82]). Moreover, it reduced the prespecified end point of end-stage kidney disease alone by 32% (HR 0.68 [95% CI 0.54–0.86]). Canagliflozin was additionally found to have a lower risk of the composite of cardiovascular death, MI, or stroke (HR 0.80 [95% CI 0.67–0.95]), as well as lower risk of hospitalizations for heart failure (HR 0.61 [95% CI 0.47–0.80]) and of the composite of cardiovascular death or hospitalization for heart failure (HR 0.69 [95% CI 0.57–0.83]). In terms of safety, no significant increase in lower-limb amputations, fractures, acute kidney injury, or hyperkalemia was noted for canagliflozin relative to placebo in CREDENCE. An increased risk for diabetic ketoacidosis was noted, however, with 2.2 and 0.2 events per 1,000 patient-years noted in the canagliflozin and placebo groups, respectively (HR 10.80 [95% CI 1.39–83.65]) (194).

The Dapagliflozin Effect on Cardiovascular Events-Thrombosis in Myocardial Infarction 58 (DECLARE-TIMI 58) trial was another randomized, double-blind trial that assessed the effects of dapagliflozin versus placebo on cardiovascular and renal outcomes in 17,160 people with type 2 diabetes and established ASCVD or multiple risk factors for ASCVD (196). Study participants had a mean age of 64 years, with ∼40% of study participants having established ASCVD at baseline—a characteristic of this trial that differs from other large cardiovascular trials where a majority of participants had established cardiovascular disease. DECLARE-TIMI 58 met the prespecified criteria for noninferiority to placebo with respect to major adverse cardiovascular events but did not show a lower rate of major adverse cardiovascular events when compared with placebo (8.8% in the dapagliflozin group and 9.4% in the placebo group; HR 0.93 [95% CI 0.84–1.03]; P = 0.17). A lower rate of cardiovascular death or hospitalization for heart failure was noted (4.9% vs. 5.8%; HR 0.83 [95% CI 0.73–0.95]; P = 0.005), which reflected a lower rate of hospitalization for heart failure (HR 0.73 [95% CI 0.61–0.88]). No difference was seen in cardiovascular death between groups.

In the Dapagliflozin and Prevention of Adverse Outcomes in Chronic Kidney Disease (DAPA-CKD) trial (197), 4,304 individuals with CKD (UACR 200–5,000 mg/g and eGFR 25–75 mL/min/1.73 m2), with or without diabetes, were randomized to dapagliflozin 10 mg daily or placebo. The primary outcome was a composite of sustained decline in eGFR of at least 50%, end-stage kidney disease, or death from renal or cardiovascular causes. Over a median follow-up period of 2.4 years, a primary outcome event occurred in 9.2% of participants in the dapagliflozin group and 14.5% of those in the placebo group. The risk of the primary composite outcome was significantly lower with dapagliflozin therapy compared with placebo (HR 0.61 [95% CI 0.51–0.72]), as were the risks for a renal composite outcome of sustained decline in eGFR of at least 50%, endstage kidney disease, or death from renal causes (HR 0.56 [95% CI 0.45–0.68]), and a composite of cardiovascular death or hospitalization for heart failure (HR 0.71 [95% CI 0.55–0.92]). The effects of dapagliflozin therapy were similar in individuals with and without type 2 diabetes.

Results of the Dapagliflozin and Prevention of Adverse Outcomes in Heart Failure (DAPA-HF) trial, the Empagliflozin Outcome Trial in Patients With Chronic Heart Failure and a Reduced Ejection Fraction (EMPEROR-Reduced), Empagliflozin Outcome Trial in Patients With Chronic Heart Failure With Preserved Ejection Fraction (EMPEROR-Preserved), Effects of Dapagliflozin on Biomarkers, Symptoms and Functional Status in Patients With PRESERVED Ejection Fraction Heart Failure (PRESERVED-HF), and Dapagliflozin Evaluation to Improve the Lives of Patients with Preserved Ejection Fraction Heart Failure (DELIVER), which assessed the effects of dapagliflozin and empagliflozin in individuals with established heart failure (11,189,198,199,200), are described below in glucose-lowering therapies and heart failure.

The Evaluation of Ertugliflozin Efficacy and Safety Cardiovascular Outcomes Trial (VERTIS CV) (201) was a randomized, double-blind trial that established the effects of ertugliflozin versus placebo on cardiovascular outcomes in 8,246 people with type 2 diabetes and established ASCVD. Participants were assigned to the addition of 5 mg or 15 mg of ertugliflozin or to placebo once daily to background standard care. Study participants had a mean age of 64.4 years and a mean duration of diabetes of 13 years at baseline and were followed for a median of 3.0 years. VERTIS CV met the prespecified criteria for noninferiority of ertugliflozin to placebo with respect to the primary outcome of major adverse cardiovascular events (11.9% in the pooled ertugliflozin group and 11.9% in the placebo group; HR 0.97 [95% CI 0.85–1.11]; P < 0.001). Ertugliflozin was not superior to placebo for the key secondary outcomes of death from cardiovascular causes or hospitalization for heart failure; death from cardiovascular causes; or the composite of death from renal causes, renal replacement therapy, or doubling of the serum creatinine level. The HR for a secondary outcome of hospitalization for heart failure (ertugliflozin vs. placebo) was 0.70 [95% CI 0.54–0.90], consistent with findings from other SGLT2 inhibitor cardiovascular outcomes trials.

Sotagliflozin, an SGLT1 and SGLT2 inhibitor not currently approved by the FDA in the U.S., lowers glucose via delayed glucose absorption in the gut in addition to increasing urinary glucose excretion and has been evaluated in the Effect of Sotagliflozin on Cardiovascular and Renal Events in Patients With Type 2 Diabetes and Moderate Renal Impairment Who Are at Cardiovascular Risk (SCORED) trial (202). A total of 10,584 people with type 2 diabetes, CKD, and additional cardiovascular risk were enrolled in SCORED and randomized to sotagliflozin 200 mg once daily (uptitrated to 400 mg once daily if tolerated) or placebo. SCORED ended early due to a lack of funding; thus, changes to the prespecified primary end points were made prior to unblinding to accommodate a lower than anticipated number of end point events. The primary end point of the trial was the total number of deaths from cardiovascular causes, hospitalizations for heart failure, and urgent visits for heart failure. After a median of 16 months of follow-up, the rate of primary end point events was reduced with sotagliflozin (5.6 events per 100 patient-years in the sotagliflozin group and 7.5 events per 100 patient-years in the placebo group [HR 0.74 (95% CI 0.63–0.88); P < 0.001]). Sotagliflozin also reduced the risk of the secondary end point of total number of hospitalizations for heart failure and urgent visits for heart failure (3.5% in the sotagliflozin group and 5.1% in the placebo group; HR 0.67 [95% CI 0.55–0.82]; P < 0.001) but not the secondary end point of deaths from cardiovascular causes. No significant between-group differences were found for the outcome of all-cause mortality or for a composite renal outcome comprising the first occurrence of long-term dialysis, renal transplantation, or a sustained reduction in eGFR. In general, the adverse effects of sotagliflozin were similar to those seen with use of SGLT2 inhibitors, but they also included an increased rate of diarrhea potentially related to the inhibition of SGLT1.

GLP-1 Receptor Agonist Trials

The Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results (LEADER) trial was a randomized, double-blind trial that assessed the effect of liraglutide, a glucagon-like peptide 1 (GLP-1) receptor agonist, versus placebo on cardiovascular outcomes in 9,340 people with type 2 diabetes at high risk for cardiovascular disease or with cardiovascular disease (203). Study participants had a mean age of 64 years and a mean duration of diabetes of nearly 13 years. Over 80% of study participants had established cardiovascular disease. After a median follow-up of 3.8 years, LEADER showed that the primary composite outcome (MI, stroke, or cardiovascular death) occurred in fewer participants in the treatment group (13.0%) when compared with the placebo group (14.9%) (HR 0.87 [95% CI 0.78–0.97]; P < 0.001 for noninferiority; P = 0.01 for superiority). Deaths from cardiovascular causes were significantly reduced in the liraglutide group (4.7%) compared with the placebo group (6.0%) (HR 0.78 [95% CI 0.66–0.93]; P = 0.007) (203).

Results from a moderate-sized trial of another GLP-1 receptor agonist, semaglutide, were consistent with the LEADER trial (204). Semaglutide is a once-weekly GLP-1 receptor agonist approved by the FDA for the treatment of type 2 diabetes. The Trial to Evaluate Cardiovascular and Other Long-term Outcomes With Semaglutide in Subjects With Type 2 Diabetes (SUSTAIN-6) was the initial randomized trial powered to test noninferiority of semaglutide for the purpose of regulatory approval (204). In this study, 3,297 people with type 2 diabetes were randomized to receive once-weekly semaglutide (0.5 mg or 1.0 mg) or placebo for 2 years. The primary outcome (the first occurrence of cardiovascular death, nonfatal MI, or nonfatal stroke) occurred in 108 patients (6.6%) in the semaglutide group vs. 146 patients (8.9%) in the placebo group (HR 0.74 [95% CI 0.58–0.95]; P < 0.001). More patients discontinued treatment in the semaglutide group because of adverse events, mainly gastrointestinal. The cardiovascular effects of the oral formulation of semaglutide compared with placebo have been assessed in Peptide Innovation for Early Diabetes Treatment (PIONEER) 6, a preapproval trial designed to rule out an unacceptable increase in cardiovascular risk (205). In this trial of 3,183 people with type 2 diabetes and high cardiovascular risk followed for a median of 15.9 months, oral semaglutide was noninferior to placebo for the primary composite outcome of cardiovascular death, nonfatal MI, or nonfatal stroke (HR 0.79 [95% CI 0.57–1.11]; P < 0.001 for noninferiority) (205). The cardiovascular effects of this formulation of semaglutide will be further tested in a large, longer-term outcomes trial.

The Harmony Outcomes trial randomized 9,463 people with type 2 diabetes and cardiovascular disease to once-weekly subcutaneous albiglutide or matching placebo, in addition to their standard care (206). Over a median duration of 1.6 years, the GLP-1 receptor agonist reduced the risk of cardiovascular death, MI, or stroke to an incidence rate of 4.6 events per 100 person-years in the albiglutide group vs. 5.9 events in the placebo group (HR ratio 0.78, P = 0.0006 for superiority) (206). This agent is not currently available for clinical use.

The Researching Cardiovascular Events With a Weekly Incretin in Diabetes (REWIND) trial was a randomized, double-blind, placebo-controlled trial that assessed the effect of the once-weekly GLP-1 receptor agonist dulaglutide versus placebo on major adverse cardiovascular events in ∼9,990 people with type 2 diabetes at risk for cardiovascular events or with a history of cardiovascular disease (207). Study participants had a mean age of 66 years and a mean duration of diabetes of ∼10 years. Approximately 32% of participants had history of atherosclerotic cardiovascular events at baseline. After a median follow-up of 5.4 years, the primary composite outcome of nonfatal MI, nonfatal stroke, or death from cardiovascular causes occurred in 12.0% and 13.4% of participants in the dulaglutide and placebo treatment groups, respectively (HR 0.88 [95% CI 0.79–0.99]; P = 0.026). These findings equated to incidence rates of 2.4 and 2.7 events per 100 person-years, respectively. The results were consistent across the subgroups of patients with and without history of CV events. Allcause mortality did not differ between groups (P = 0.067).

The Evaluation of Lixisenatide in Acute Coronary Syndrome (ELIXA) trial studied the once-daily GLP-1 receptor agonist lixisenatide on cardiovascular outcomes in people with type 2 diabetes who had had a recent acute coronary event (208). A total of 6,068 people with type 2 diabetes with a recent hospitalization for MI or unstable angina within the previous 180 days were randomized to receive lixisenatide or placebo in addition to standard care and were followed for a median of ∼2.1 years. The primary outcome of cardiovascular death, MI, stroke, or hospitalization for unstable angina occurred in 406 patients (13.4%) in the lixisenatide group vs. 399 (13.2%) in the placebo group (HR 1.2 [95% CI 0.89–1.17]), which demonstrated the noninferiority of lixisenatide to placebo (P < 0.001) but did not show superiority (P = 0.81).

The Exenatide Study of Cardiovascular Event Lowering (EXSCEL) trial also reported results with the once-weekly GLP-1 receptor agonist extended-release exenatide and found that major adverse cardiovascular events were numerically lower with use of extended-release exenatide compared with placebo, although this difference was not statistically significant (209). A total of 14,752 people with type 2 diabetes (of whom 10,782 [73.1%] had previous cardiovascular disease) were randomized to receive extended-release exenatide 2 mg or placebo and followed for a median of 3.2 years. The primary end point of cardiovascular death, MI, or stroke occurred in 839 patients (11.4%; 3.7 events per 100 person-years) in the exenatide group and in 905 patients (12.2%; 4.0 events per 100 person-years) in the placebo group (HR 0.91 [95% CI 0.83–1.00]; P < 0.001 for noninferiority), but exenatide was not superior to placebo with respect to the primary end point (P = 0.06 for superiority). However, all-cause mortality was lower in the exenatide group (HR 0.86 [95% CI 0.77–0.97]). The incidence of acute pancreatitis, pancreatic cancer, medullary thyroid carcinoma, and serious adverse events did not differ significantly between the two groups.

In summary, there are now numerous large randomized controlled trials reporting statistically significant reductions in cardiovascular events for three of the FDA-approved SGLT2 inhibitors (empagliflozin, canagliflozin, dapagliflozin, with lesser benefits seen with ertugliflozin) and four FDA-approved GLP-1 receptor agonists (liraglutide, albiglutide [although that agent was removed from the market for business reasons], semaglutide [lower risk of cardiovascular events in a moderate-sized clinical trial but one not powered as a cardiovascular outcomes trial], and dulaglutide). Meta-analyses of the trials reported to date suggest that GLP-1 receptor agonists and SGLT2 inhibitors reduce risk of atherosclerotic major adverse cardiovascular events to a comparable degree in people with type 2 diabetes and established ASCVD (210,211). SGLT2 inhibitors also reduce risk of heart failure hospitalization and progression of kidney disease in people with established ASCVD, multiple risk factors for ASCVD, or albuminuric kidney disease (212,213). In people with type 2 diabetes and established ASCVD, multiple ASCVD risk factors, or diabetic kidney disease, an SGLT2 inhibitor with demonstrated cardiovascular benefit is recommended to reduce the risk of major adverse cardiovascular events and/or heart failure hospitalization. In people with type 2 diabetes and established ASCVD or multiple risk factors for ASCVD, a glucagon-like peptide 1 receptor agonist with demonstrated cardiovascular benefit is recommended to reduce the risk of major adverse cardiovascular events. For many patients, use of either an SGLT2 inhibitor or a GLP-1 receptor agonist to reduce cardiovascular risk is appropriate. Emerging data suggest that use of both classes of drugs will provide an additive cardiovascular and kidney outcomes benefit; thus, combination therapy with an SGLT2 inhibitor and a GLP-1 receptor agonist may be considered to provide the complementary outcomes benefits associated with these classes of medication. Evidence to support such an approach includes findings from AMPLITUDE-O (Effect of Efpeglenatide on Cardiovascular Outcomes), an outcomes trial of people with type 2 diabetes and either cardiovascular or kidney disease plus at least one other risk factor randomized to the investigational GLP-1 receptor agonist efpeglenatide or placebo (214). Randomization was stratified by current or potential use of SGLT2 inhibitor therapy, a class ultimately used by >15% of the trial participants. Over a median follow-up of 1.8 years, efpeglenatide therapy reduced the risk of incident major adverse cardiovascular events by 27% and of a composite renal outcome event by 32%. Importantly, the effects of efpeglenatide did not vary by use of SGLT2 inhibitors, suggesting that the beneficial effects of the GLP-1 receptor agonist were independent of those provided by SGLT2 inhibitor therapy (215). Efpeglenatide is currently not approved by the FDA for use in the U.S.

Glucose-Lowering Therapies and Heart Failure

As many as 50% of people with type 2 diabetes may develop heart failure (216). These conditions, which are each associated with increased morbidity and mortality, commonly coincide, and independently contribute to adverse outcomes (217). Strategies to mitigate these risks are needed, and the heart failure-related risks and benefits of glucose-lowering medications should be considered carefully when determining a regimen of care for people with diabetes and either established heart failure or high risk for the development of heart failure.

Data on the effects of glucose-lowering agents on heart failure outcomes have demonstrated that thiazolidinediones have a strong and consistent relationship with increased risk of heart failure (218220). Therefore, thiazolidinedione use should be avoided in people with symptomatic heart failure. Restrictions to use of metformin in people with medically treated heart failure were removed by the FDA in 2006 (221). Observational studies of people with type 2 diabetes and heart failure suggest that metformin users have better outcomes than individuals treated with other antihyperglycemic agents (222); however, no randomized trial of metformin therapy has been conducted in people with heart failure. Metformin may be used for the management of hyperglycemia in people with stable heart failure as long as kidney function remains within the recommended range for use (223).

Recent studies examining the relationship between DPP-4 inhibitors and heart failure have had mixed results. The Saxagliptin Assessment of Vascular Outcomes Recorded in Patients with Diabetes Mellitus – Thrombolysis in Myocardial Infarction 53 (SAVOR-TIMI 53) study showed that patients treated with the DPP-4 inhibitor saxagliptin were more likely to be hospitalized for heart failure than those given placebo (3.5% vs. 2.8%, respectively) (224). However, three other cardiovascular outcomes trials—Examination of Cardiovascular Outcomes with Alogliptin versus Standard of Care (EXAMINE) (225), Trial Evaluating Cardiovascular Outcomes with Sitagliptin (TECOS) (226), and the Cardiovascular and Renal Microvascular Outcome Study With Linagliptin (CARMELINA) (193)—did not find a significant increase in risk of heart failure hospitalization with DPP-4 inhibitor use compared with placebo. No increased risk of heart failure hospitalization has been identified in the cardiovascular outcomes trials of the GLP-1 receptor agonists lixisenatide, liraglutide, semaglutide, exenatide once-weekly, albiglutide, or dulaglutide compared with placebo (Table 10.3B) (203,204,207209).

Reduced incidence of heart failure has been observed with the use of SGLT2 inhibitors (8,194,196). In EMPA- REG OUTCOME, the addition of empagliflozin to standard care led to a significant 35% reduction in hospitalization for heart failure compared with placebo (8). Although the majority of patients in the study did not have heart failure at baseline, this benefit was consistent in patients with and without a history of heart failure (10). Similarly, in CANVAS and DECLARE-TIMI 58, there were 33% and 27% reductions in hospitalization for heart failure, respectively, with SGLT2 inhibitor use versus placebo (9,196). Additional data from the CREDENCE trial with canagliflozin showed a 39% reduction in hospitalization for heart failure, and 31% reduction in the composite of cardiovascular death or hospitalization for heart failure, in a diabetic kidney disease population with albuminuria (UACR >300 to 5,000 mg/g) (194). These combined findings from four large outcomes trials of three different SGLT2 inhibitors are highly consistent and clearly indicate robust benefits of SGLT2 inhibitors in the prevention of heart failure hospitalizations. The EMPA-REG OUTCOME, CANVAS, DECLARE-TIMI 58, and CREDENCE trials suggested, but did not prove, that SGLT2 inhibitors would be beneficial in the treatment of people with established heart failure. More recently, the placebo-controlled DAPA-HF trial evaluated the effects of dapagliflozin on the primary outcome of a composite of worsening heart failure or cardiovascular death in patients with New York Heart Association (NYHA) class II, III, or IV heart failure and an ejection fraction of 40% or less. Of the 4,744 trial participants, 45% had a history of type 2 diabetes. Over a median of 18.2 months, the group assigned to dapagliflozin treatment had a lower risk of the primary outcome (HR 0.74 [95% CI 0.65–0.85]), lower risk of first worsening heart failure event (HR 0.70 [95% CI 0.59–0.83]), and lower risk of cardiovascular death (HR 0.82 [95% CI 0.69–0.98]) compared with placebo. The effect of dapagliflozin on the primary outcome was consistent regardless of the presence or absence of type 2 diabetes (11).

EMPEROR-Reduced assessed the effects of empagliflozin 10 mg once daily versus placebo on a primary composite outcome of cardiovascular death or hospitalization for worsening heart failure in a population of 3,730 patients with NYHA class II, III, or IV heart failure and an ejection fraction of 40% or less (200). At baseline, 49.8% of participants had a history of diabetes. Over a median follow-up of 16 months, those in the empagliflozin-treated group had a reduced risk of the primary outcome (HR 0.75 [95% CI 0.65–0.86]; P < 0.001) and fewer total hospitalizations for heart failure (HR 0.70 [95% CI 0.58–0.85]; P < 0.001). The effect of empagliflozin on the primary outcome was consistent irrespective of diabetes diagnosis at baseline. The risk of a prespecified renal composite outcome (chronic dialysis, renal transplantation, or a sustained reduction in eGFR) was lower in the empagliflozin group than in the placebo group (1.6% in the empagliflozin group vs. 3.1% in the placebo group; HR 0.50 [95% CI 0.32–0.77]).

EMPEROR-Preserved, a randomized double-blinded placebo-controlled trial of 5,988 adults with NYHA functional class I–IV chronic HFpEF (left ventricular ejection fraction >40%), evaluated the efficacy of empagliflozin 10 mg daily versus placebo on top of standard of care on the primary outcome of composite cardiovascular death or hospitalization for heart failure (189). Approximately 50% of subjects had type 2 diabetes at baseline. Over a median of 26.2 months, there was a 21% reduction (HR 0.79 [95% CI 0.69–0.90]; P < 0.001) of the primary outcome. The effects of empagliflozin were consistent in people with or without diabetes (189).

In the DELIVER trial, 6,263 individuals with heart failure and an ejection fraction >40% were randomized to receive either dapagliflozin or placebo (199). The primary outcome of a composite of worsening heart failure, defined as hospitalization or urgent visit for heart failure, or cardiovascular death was reduced by 18% in patients treated with dapagliflozin compared with placebo (HR 0.82 [95% CI 0.73–0.92]; P < 0.001). Approximately 44% of patients randomized to either dapagliflozin or placebo had type 2 diabetes, and results were consistent regardless of the presence of type 2 diabetes.

A large recent meta-analysis (227) including data from EMPEROR-Reduced, EMPEROR-Preserved, DAPA-HF, DELIVER, and Effect of Sotagliflozin on Cardiovascular Events in Patients With Type 2 Diabetes Post Worsening Heart Failure (SOLOIST-WHF) included 21,947 patients and demonstrated reduced risk for the composite of cardiovascular death or hospitalization for heart failure, cardiovascular death, first hospitalization for heart failure, and all-cause mortality. The findings on the studied end points were consistent in both trials of heart failure with mildly reduced or preserved ejection fraction and in all five trials combined. Collectively, these studies indicate that SGLT2 inhibitors reduce the risk for heart failure hospitalization and cardiovascular death in a wide range of people with heart failure.

Additional data are accumulating regarding the effects of SGLT inhibition in people hospitalized for acute decompensated heart failure and in people with heart failure and HFpEF. As an example, the investigational SGLT1 and SGLT2 inhibitor sotagliflozin has also been studied in the SOLOIST-WHF trial (228). In SOLOIST-WHF, 1,222 people with type 2 diabetes who were recently hospitalized for worsening heart failure were randomized to sotagliflozin 200 mg once daily (with uptitration to 400 mg once daily if tolerated) or placebo either before or within 3 days after hospital discharge. Patients were eligible if hospitalized for signs and symptoms of heart failure (including elevated natriuretic peptide levels) requiring treatment with intravenous diuretic therapy. Exclusion criteria included end-stage heart failure or recent acute coronary syndrome or intervention, or an eGFR <30 mL/min/1.73 m2). Patients were required to be clinically stable prior to randomization, defined as no use of supplemental oxygen, a systolic blood pressure ≥100 mmHg, and no need for intravenous inotropic or vasodilator therapy other than nitrates. Similar to SCORED, SOLOIST-WHF ended early due to a lack of funding, resulting in a change to the prespecified primary end point prior to unblinding to accommodate a lower than anticipated number of end point events. At a median follow-up of 9 months, the rate of primary end point events (the total number of cardiovascular deaths and hospitalizations and urgent visits for heart failure) was lower in the sotagliflozin group than in the placebo group (51.0 vs. 76.3; HR 0.67 [95% CI 0.52–0.85]; P < 0.001). No significant between-group differences were found in the rates of cardiovascular death or all-cause mortality. Both diarrhea (6.1% vs. 3.4%) and severe hypoglycemia (1.5% vs. 0.3%) were more common with sotagliflozin than with placebo. The trial was originally also intended to evaluate the effects of SGLT inhibition in people with HFpEF, and ultimately no evidence of heterogeneity of treatment effect by ejection fraction was noted. However, the relatively small percentage of such patients enrolled (only 21% of participants had ejection fraction >50%) and the early termination of the trial limited the ability to determine the effects of sotagliflozin in HFpEF specifically.

In addition to the hospitalization and mortality benefit in people with heart failure, several recent analyses have addressed whether SGLT2 inhibitor treatment improves clinical stability and functional status in individuals with heart failure. In 3,730 patients with NYHA class II–IV heart failure with an ejection fraction of ≤40%, treatment with empagliflozin reduced the combined risk of death, hospitalization for heart failure, or an emergent/urgent heart failure visit requiring intravenous treatment and reduced the total number of hospitalizations for heart failure requiring intensive care, a vasopressor or positive inotropic drug, or mechanical or surgical intervention (229). In addition, patients treated with empagliflozin were more likely to experience an improvement in NYHA functional class (229). In people hospitalized for acute de novo or decompensated chronic heart failure, initiation of empagliflozin treatment during hospitalization reduced the primary outcome of a composite of death from any cause, number of heart failure events and time to first heart failure event, or a 5-point or greater difference in change from baseline in the Kansas City Cardiomyopathy Questionnaire Total Symptom Score (230). Furthermore, PRESERVED-HF, a multicenter study (26 sites in the U.S.) showed that dapagliflozin treatment leads to significant improvement in both symptoms and physical limitation, as well as objective measures of exercise function in people with chronic HFpEF, regardless of diabetes status (198). Finally, canagliflozin improved heart failure symptoms assessed using the Kansas City Cardiomyopathy Questionnaire Total Symptom Score, irrespective of left ventricular ejection fraction or the presence of diabetes (231). Therefore, in people with type 2 diabetes and established HFpEF or HFrEF, an SGLT2 inhibitor with proven benefit in this patient population is recommended to reduce the risk of worsening heart failure and cardiovascular death. In addition, an SGLT2 inhibitor is recommended in this patient population to improve symptoms, physical limitations, and quality of life. The benefits seen in this patient population likely represent a class effect, and they appear unrelated to glucose lowering given comparable outcomes in people with heart failure with and without diabetes.

Finerenone in People with Type 2 Diabetes and Chronic Kidney Disease

As discussed in detail in Section 11, “Chronic Kidney Disease and Risk Management,” people with diabetes are at an increased risk for CKD, which increases cardiovascular risk (232). Finerenone, a selective nonsteroidal mineralocorticoid antagonist, has been shown in the Finerenone in Reducing Kidney Failure and Disease Progression in Diabetic Kidney Disease (FIDELIO-DKD) trial to improve CKD outcomes in people with type 2 diabetes with stage 3 or 4 CKD and severe albuminuria (233). In the Finerenone in Reducing Cardiovascular Mortality and Morbidity in Diabetic Kidney Disease (FIGARO-DKD) trial, 7,437 patients with UACR 30–300 mg/g and eGFR 25–90 mL/min/1.73 m2 or UACR 300–5,000 and eGFR ≥60 mL/min/1.73 m2 on maximum dose of renin-angiotensin system blockade were randomized to receive finerenone or placebo (186). The HR of the primary outcome of cardiovascular death, nonfatal MI, nonfatal stroke, or hospitalization from heart failure was reduced by 13% in patients treated with finerenone. A prespecified subgroup analysis from FIGARO-DKD further revealed that in patients without symptomatic HFrEF, finerenone reduces the risk for new-onset heart failure and improves heart failure outcomes in people with type 2 diabetes and CKD (187). Finally, in the pooled analysis of 13,026 people with type 2 diabetes and CKD from both FIDELIO-DKD and FIGARO-DKD, the HRs for the composite of cardiovascular death, nonfatal MI, nonfatal stroke, or hospitalization for heart failure as well as a composite of kidney failure, a sustained ≥57% decrease in eGFR from baseline over ≥4 weeks, or renal death were 0.86 and 0.77, respectively (188). These collective studies indicate that finerenone improves cardiovascular and renal outcomes in people with type 2 diabetes. Therefore, in people with type 2 diabetes and CKD with albuminuria treated with maximum tolerated doses of ACE inhibitor or ARB, addition of finernone should be considered to improve cardiovascular outcomes and reduce the risk of CKD progression.

Clinical Approach

As has been carefully outlined in Fig. 9.3 in the preceding Section 9, “Pharmacologic Approaches to Glycemic Treatment,” people with type 2 diabetes with or at high risk for ASCVD, heart failure, or CKD should be treated with a cardioprotective SGLT2 inhibitor and/or GLP-1 receptor agonist as part of the comprehensive approach to cardiovascular and kidney risk reduction. Importantly, these agents should be included in the regimen of care irrespective of the need for additional glucose lowering, and irrespective of metformin use. Such an approach has also been described in the American Diabetes Association–endorsed American College of Cardiology “2020 Expert Consensus Decision Pathway on Novel Therapies for Cardiovascular Risk Reduction in Patients With Type 2 Diabetes” (234). Figure 10.3, reproduced from that decision pathway, outlines the approach to risk reduction with SGLT2 inhibitor or GLP-1 receptor agonist therapy in conjunction with other traditional, guideline-based preventive medical therapies for blood pressure, lipids, and glycemia and antiplatelet therapy.

Figure 10.3.

Figure 10.3

Approach to risk reduction with SGLT2 inhibitor or GLP-1 receptor agonist therapy in conjunction with other traditional, guideline-based preventive medical therapies for blood pressure, lipids, and glycemia and antiplatelet therapy. Reprinted with permission from Das et al. (234).

Adoption of these agents should be reasonably straightforward in people with established cardiovascular or kidney disease who are later diagnosed with diabetes, as the cardioprotective agents can be used from the outset of diabetes management. On the other hand, incorporation of SGLT2 inhibitor or GLP-1 receptor agonist therapy in the care of individuals with more long-standing diabetes may be more challenging, particularly if patients are using an already complex glucose-lowering regimen. In such patients, SGLT2 inhibitor or GLP-1 receptor agonist therapy may need to replace some or all of their existing medications to minimize risks of hypoglycemia and adverse side effects, and potentially to minimize medication costs. Close collaboration between primary and specialty care professionals can help to facilitate these transitions in clinical care and, in turn, improve outcomes for highrisk people with type 2 diabetes.

Footnotes

Disclosure information for each author is available at https://doi.org/10.2337/dc23-SDIS.

This section has received endorsement from the American College of Cardiology.

Suggested citation: ElSayed NA, Aleppo G, Aroda VR, et al., American Diabetes Association. 10. Cardiovascular disease and risk management: Standards of Care in Diabetes—2023. Diabetes Care 2023;46(Suppl. 1):S158–S190

References

  • 1. American Diabetes Association . Economic costs of diabetes in the U.S. in 2017. Diabetes Care 2018;41:917–928 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Ali MK, Bullard KM, Saaddine JB, Cowie CC, Imperatore G, Gregg EW. Achievement of goals in U.S. diabetes care, 1999-2010. N Engl J Med 2013;368:1613–1624 [DOI] [PubMed] [Google Scholar]
  • 3. Buse JB, Ginsberg HN, Bakris GL, et al.; American Heart Association; American Diabetes Association . 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;30:162–172 [DOI] [PubMed] [Google Scholar]
  • 4. Gaede P, Lund-Andersen H, Parving HH, Pedersen O. Effect of a multifactorial intervention on mortality in type 2 diabetes. N Engl J Med 2008;358:580–591 [DOI] [PubMed] [Google Scholar]
  • 5. Cavender MA, Steg PG, Smith SC Jr, et al.; REACH Registry Investigators . Impact of diabetes mellitus on hospitalization for heart failure, cardiovascular events, and death: outcomes at 4 years from the Reduction of Atherothrombosis for Continued Health (REACH) Registry. Circulation 2015;132:923–931 [DOI] [PubMed] [Google Scholar]
  • 6. McAllister DA, Read SH, Kerssens J, et al. Incidence of hospitalization for heart failure and case-fatality among 3.25 million people with and without diabetes mellitus. Circulation 2018;138:2774–2786 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Lam CSP, Voors AA, de Boer RA, Solomon SD, van Veldhuisen DJ. Heart failure with preserved ejection fraction: from mechanisms to therapies. Eur Heart J 2018;39:2780–2792 [DOI] [PubMed] [Google Scholar]
  • 8. Zinman B, Wanner C, Lachin JM, et al.; EMPA-REG OUTCOME Investigators . Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 2015;373:2117–2128 [DOI] [PubMed] [Google Scholar]
  • 9. Neal B, Perkovic V, Mahaffey KW, et al.; CANVAS Program Collaborative Group . Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med 2017;377:644–657 [DOI] [PubMed] [Google Scholar]
  • 10. Fitchett D, Butler J, van de Borne P, et al.; EMPA-REG OUTCOME trial investigators . Effects of empagliflozin on risk for cardiovascular death and heart failure hospitalization across the spectrum of heart failure risk in the EMPA-REG OUTCOME trial. Eur Heart J 2018;39:363–370 [DOI] [PubMed] [Google Scholar]
  • 11. McMurray JJV, Solomon SD, Inzucchi SE, et al.; DAPA-HF Trial Committees and Investigators . Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med 2019;381:1995–2008 [DOI] [PubMed] [Google Scholar]
  • 12. Arnott C, Li Q, Kang A, et al. Sodium-glucose cotransporter 2 inhibition for the prevention of cardiovascular events in patients with type 2 diabetes mellitus: a systematic review and meta-analysis. J Am Heart Assoc 2020;9:e014908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Blood Pressure Lowering Treatment Trialists’ Collaboration . Blood pressure-lowering treatment based on cardiovascular risk: a meta-analysis of individual patient data. Lancet 2014;384:591–598 [DOI] [PubMed] [Google Scholar]
  • 14. Grundy SM, Stone NJ, Bailey AL, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol 2019;73:3168–3209 [DOI] [PubMed] [Google Scholar]
  • 15. Muntner P, Colantonio LD, Cushman M, et al. Validation of the atherosclerotic cardiovascular disease Pooled Cohort risk equations. JAMA 2014;311:1406–1415 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. DeFilippis AP, Young R, McEvoy JW, et al. Risk score overestimation: the impact of individual cardiovascular risk factors and preventive therapies on the performance of the American Heart Association-American College of Cardiology-Atherosclerotic Cardiovascular Disease risk score in a modern multi-ethnic cohort. Eur Heart J 2017;38:598–608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Bohula EA, Morrow DA, Giugliano RP, et al. Atherothrombotic risk stratification and ezetimibe for secondary prevention. J Am Coll Cardiol 2017;69:911–921 [DOI] [PubMed] [Google Scholar]
  • 18. Bohula EA, Bonaca MP, Braunwald E, et al. Atherothrombotic risk stratification and the efficacy and safety of vorapaxar in patients with stable ischemic heart disease and previous myocardial infarction. Circulation 2016;134:304–313 [DOI] [PubMed] [Google Scholar]
  • 19. Whelton PK, Carey RM, Aronow WS, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. J Am Coll Cardiol 2018;71:e127–e248 [DOI] [PubMed] [Google Scholar]
  • 20. de Boer IH, Bangalore S, Benetos A, et al. Diabetes and hypertension: a position statement by the American Diabetes Association. Diabetes Care 2017;40:1273–1284 [DOI] [PubMed] [Google Scholar]
  • 21. Unger T, Borghi C, Charchar F, et al. 2020 International Society of Hypertension Global Hypertension Practice Guidelines. Hypertension 2020;75:1334–1357 [DOI] [PubMed] [Google Scholar]
  • 22. Williams B, Mancia G, Spiering W, et al.; ESC Scientific Document Group . 2018 ESC/ESH guidelines for the management of arterial hypertension. Eur Heart J 2018;39:3021–3104 [DOI] [PubMed] [Google Scholar]
  • 23. Bobrie G, Genès N, Vaur L, et al. Is “isolated home” hypertension as opposed to “isolated office” hypertension a sign of greater cardiovascular risk? Arch Intern Med 2001;161:2205–2211 [DOI] [PubMed] [Google Scholar]
  • 24. Sega R, Facchetti R, Bombelli M, et al. Prognostic value of ambulatory and home blood pressures compared with office blood pressure in the general population: follow-up results from the Pressioni Arteriose Monitorate e Loro Associazioni (PAMELA) study. Circulation 2005;111:1777–1783 [DOI] [PubMed] [Google Scholar]
  • 25. Omboni S, Gazzola T, Carabelli G, Parati G. Clinical usefulness and cost effectiveness of home blood pressure telemonitoring: meta-analysis of randomized controlled studies. J Hypertens 2013;31:455–467; discussion 467–468 [DOI] [PubMed] [Google Scholar]
  • 26. Emdin CA, Rahimi K, Neal B, Callender T, Perkovic V, Patel A. Blood pressure lowering in type 2 diabetes: a systematic review and meta-analysis. JAMA 2015;313:603–615 [DOI] [PubMed] [Google Scholar]
  • 27. Arguedas JA, Leiva V, Wright JM. Blood pressure targets for hypertension in people with diabetes mellitus. Cochrane Database Syst Rev 2013;10:CD008277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Ettehad D, Emdin CA, Kiran A, et al. Blood pressure lowering for prevention of cardiovascular disease and death: a systematic review and meta-analysis. Lancet 2016;387:957–967 [DOI] [PubMed] [Google Scholar]
  • 29. Brunström M, Carlberg B. Effect of antihypertensive treatment at different blood pressure levels in patients with diabetes mellitus: systematic review and meta-analyses. BMJ 2016;352:i717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Bangalore S, Kumar S, Lobach I, Messerli FH. Blood pressure targets in subjects with type 2 diabetes mellitus/impaired fasting glucose: observations from traditional and bayesian random-effects meta-analyses of randomized trials. Circulation 2011;123:2799–2810 [DOI] [PubMed] [Google Scholar]
  • 31. Thomopoulos C, Parati G, Zanchetti A. Effects of blood-pressure-lowering treatment on outcome incidence in hypertension: 10 - Should blood pressure management differ in hypertensive patients with and without diabetes mellitus? Overview and meta-analyses of randomized trials. J Hypertens 2017;35:922–944 [DOI] [PubMed] [Google Scholar]
  • 32. Xie X, Atkins E, Lv J, et al. Effects of intensive blood pressure lowering on cardiovascular and renal outcomes: updated systematic review and meta-analysis. Lancet 2016;387:435–443 [DOI] [PubMed] [Google Scholar]
  • 33. Wright JT Jr, Williamson JD, Whelton PK, et al.; SPRINT Research Group . A randomized trial of intensive versus standard blood-pressure control. N Engl J Med 2015;373:2103–2116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Zhang W, Zhang S, Deng Y, et al.; STEP Study Group . Trial of intensive blood-pressure control in older patients with hypertension. N Engl J Med 2021;385:1268–1279 [DOI] [PubMed] [Google Scholar]
  • 35. Cushman WC, Evans GW, Byington RP, et al.; ACCORD Study Group . Effects of intensive blood-pressure control in type 2 diabetes mellitus. N Engl J Med 2010;362:1575–1585 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Patel A, MacMahon S, Chalmers J, et al.; ADVANCE Collaborative Group . Effects of a fixed combination of perindopril and indapamide on macrovascular and microvascular outcomes in patients with type 2 diabetes mellitus (the ADVANCE trial): a randomised controlled trial. Lancet 2007;370:829–840 [DOI] [PubMed] [Google Scholar]
  • 37. Hansson L, Zanchetti A, Carruthers SG, et al.; HOT Study Group . Effects of intensive blood-pressure lowering and low-dose aspirin in patients with hypertension: principal results of the Hypertension Optimal Treatment (HOT) randomised trial. Lancet 1998;351:1755–1762 [DOI] [PubMed] [Google Scholar]
  • 38. Reboldi G, Gentile G, Angeli F, Ambrosio G, Mancia G, Verdecchia P. Effects of intensive blood pressure reduction on myocardial infarction and stroke in diabetes: a meta-analysis in 73,913 patients. J Hypertens 2011;29:1253–1269 [DOI] [PubMed] [Google Scholar]
  • 39. de Boer IH, Bakris G, Cannon CP. Individualizing blood pressure targets for people with diabetes and hypertension: comparing the ADA and the ACC/AHA recommendations. JAMA 2018;319:1319–1320 [DOI] [PubMed] [Google Scholar]
  • 40. Basu S, Sussman JB, Rigdon J, Steimle L, Denton BT, Hayward RA. Benefit and harm of intensive blood pressure treatment: derivation and validation of risk models using data from the SPRINT and ACCORD trials. PLoS Med 2017;14:e1002410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Phillips RA, Xu J, Peterson LE, Arnold RM, Diamond JA, Schussheim AE. Impact of cardiovascular risk on the relative benefit and harm of intensive treatment of hypertension. J Am Coll Cardiol 2018;71:1601–1610 [DOI] [PubMed] [Google Scholar]
  • 42. Beddhu S, Greene T, Boucher R, et al. Intensive systolic blood pressure control and incident chronic kidney disease in people with and without diabetes mellitus: secondary analyses of two randomised controlled trials. Lancet Diabetes Endocrinol 2018;6:555–563 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Sink KM, Evans GW, Shorr RI, et al. Syncope, hypotension, and falls in the treatment of hypertension: results from the randomized clinical systolic blood pressure intervention trial. J Am Geriatr Soc 2018;66:679–686 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Ilkun OL, Greene T, Cheung AK, et al. The influence of baseline diastolic blood pressure on the effects of intensive blood pressure lowering on cardiovascular outcomes and all-cause mortality in type 2 diabetes. Diabetes Care 2020;43:1878–1884 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Abalos E, Duley L, Steyn DW. Antihypertensive drug therapy for mild to moderate hypertension during pregnancy. Cochrane Database Syst Rev 2014;2:CD002252. [DOI] [PubMed] [Google Scholar]
  • 46. Magee LA, von Dadelszen P, Rey E, et al. Less-tight versus tight control of hypertension in pregnancy. N Engl J Med 2015;372:407–417 [DOI] [PubMed] [Google Scholar]
  • 47. Brown MA, Magee LA, Kenny LC, et al.; International Society for the Study of Hypertension in Pregnancy (ISSHP) . Hypertensive disorders of pregnancy: ISSHP classification, diagnosis, and management recommendations for international practice. Hypertension 2018;72:24–43 [DOI] [PubMed] [Google Scholar]
  • 48. Tita AT, Szychowski JM, Boggess K, et al.; Chronic Hypertension and Pregnancy (CHAP) Trial Consortium . Treatment for mild chronic hypertension during pregnancy. N Engl J Med 2022;386:1781–1792 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. American College of Obstetricians and Gynecologists, Task Force on Hypertension in Pregnancy . Hypertension in pregnancy. Report of the American College of Obstetricians and Gynecologists’ Task Force on Hypertension in Pregnancy. Obstet Gynecol 2013;122:1122–1131 [DOI] [PubMed] [Google Scholar]
  • 50. Al-Balas M, Bozzo P, Einarson A. Use of diuretics during pregnancy. Can Fam Physician 2009;55:44–45 [PMC free article] [PubMed] [Google Scholar]
  • 51. Irgens HU, Reisaeter L, Irgens LM, Lie RT. Long term mortality of mothers and fathers after pre-eclampsia: population based cohort study. BMJ 2001;323:1213–1217 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Sacks FM, Svetkey LP, Vollmer WM, et al.; DASH-Sodium Collaborative Research Group . Effects on blood pressure of reduced dietary sodium and the Dietary Approaches to Stop Hypertension (DASH) diet. N Engl J Med 2001;344:3–10 [DOI] [PubMed] [Google Scholar]
  • 53. James PA, Oparil S, Carter BL, et al. 2014 evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the Eighth Joint National Committee (JNC 8). JAMA 2014;311:507–520 [DOI] [PubMed] [Google Scholar]
  • 54. Mao Y, Lin W, Wen J, Chen G. Impact and efficacy of mobile health intervention in the management of diabetes and hypertension: a systematic review and meta-analysis. BMJ Open Diabetes Res Care 2020;8:e001225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Stogios N, Kaur B, Huszti E, Vasanthan J, Nolan RP. Advancing digital health interventions as a clinically applied science for blood pressure reduction: a systematic review and meta-analysis. Can J Cardiol 2020;36:764–774 [DOI] [PubMed] [Google Scholar]
  • 56. Bakris GL; Weir MR; Study of Hypertension and the Efficacy of Lotrel in Diabetes (SHIELD) Investigators . Achieving goal blood pressure in patients with type 2 diabetes: conventional versus fixed-dose combination approaches. J Clin Hypertens (Greenwich) 2003;5:202–209 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Feldman RD, Zou GY, Vandervoort MK, Wong CJ, Nelson SAE, Feagan BG. A simplified approach to the treatment of uncomplicated hypertension: a cluster randomized, controlled trial. Hypertension 2009;53:646–653 [DOI] [PubMed] [Google Scholar]
  • 58. Webster R, Salam A, de Silva HA, et al.; TRIUMPH Study Group . Fixed low-dose triple combination antihypertensive medication vs usual care for blood pressure control in patients with mild to moderate hypertension in Sri Lanka: a randomized clinical trial. JAMA 2018;320:566–579 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Bangalore S, Kamalakkannan G, Parkar S, Messerli FH. Fixed-dose combinations improve medication compliance: a meta-analysis. Am J Med 2007;120:713–719 [DOI] [PubMed] [Google Scholar]
  • 60. Catalá-López F, Macías Saint-Gerons D, González-Bermejo D, et al. Cardiovascular and renal outcomes of renin-angiotensin system blockade in adult patients with diabetes mellitus: a systematic review with network meta-analyses. PLoS Med 2016;13:e1001971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Palmer SC, Mavridis D, Navarese E, et al. Comparative efficacy and safety of blood pressure-lowering agents in adults with diabetes and kidney disease: a network meta-analysis. Lancet 2015;385:2047–2056 [DOI] [PubMed] [Google Scholar]
  • 62. Barzilay JI, Davis BR, Bettencourt J, et al.; ALLHAT Collaborative Research Group . Cardiovascular outcomes using doxazosin vs. chlorthalidone for the treatment of hypertension in older adults with and without glucose disorders: a report from the ALLHAT study. J Clin Hypertens (Greenwich) 2004;6:116–125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Weber MA, Bakris GL, Jamerson K, et al.; ACCOMPLISH Investigators . Cardiovascular events during differing hypertension therapies in patients with diabetes. J Am Coll Cardiol 2010;56:77–85 [DOI] [PubMed] [Google Scholar]
  • 64. Heart Outcomes Prevention Evaluation Study Investigators . Effects of ramipril on cardiovascular and microvascular outcomes in people with diabetes mellitus: results of the HOPE study and MICRO-HOPE substudy. Lancet 2000;355:253–259 [PubMed] [Google Scholar]
  • 65. Arnold SV, Bhatt DL, Barsness GW, et al.; American Heart Association Council on Lifestyle and Cardiometabolic Health and Council on Clinical Cardiology . Clinical management of stable coronary artery disease in patients with type 2 diabetes mellitus: a scientific statement from the American Heart Association. Circulation 2020;141:e779–e806 [DOI] [PubMed] [Google Scholar]
  • 66. Yusuf S, Teo K, Anderson C, et al.; Telmisartan Randomised AssessmeNt Study in ACE iNtolerant subjects with cardiovascular Disease (TRANSCEND) Investigators . Effects of the angiotensin-receptor blocker telmisartan on cardiovascular events in high-risk patients intolerant to angiotensin-converting enzyme inhibitors: a randomised controlled trial. Lancet 2008;372:1174–1183 [DOI] [PubMed] [Google Scholar]
  • 67. Qiao Y, Shin JI, Chen TK, et al. Association between renin-angiotensin system blockade discontinuation and all-cause mortality among persons with low estimated glomerular filtration rate. JAMA Intern Med 2020;180:718–726 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Bangalore S, Fakheri R, Toklu B, Messerli FH. Diabetes mellitus as a compelling indication for use of renin angiotensin system blockers: systematic review and meta-analysis of randomized trials. BMJ 2016;352:i438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Carlberg B, Samuelsson O, Lindholm LH. Atenolol in hypertension: is it a wise choice? Lancet 2004;364:1684–1689 [DOI] [PubMed] [Google Scholar]
  • 70. Murphy SP, Ibrahim NE, Januzzi JL Jr. Heart failure with reduced ejection fraction: a review. JAMA 2020;324:488–504 [DOI] [PubMed] [Google Scholar]
  • 71. Yusuf S, Teo KK, Pogue J, et al.; ONTARGET Investigators . Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med 2008;358:1547–1559 [DOI] [PubMed] [Google Scholar]
  • 72. Fried LF, Emanuele N, Zhang JH, et al.; VA NEPHRON-D Investigators . Combined angiotensin inhibition for the treatment of diabetic nephropathy. N Engl J Med 2013;369:1892–1903 [DOI] [PubMed] [Google Scholar]
  • 73. Makani H, Bangalore S, Desouza KA, Shah A, Messerli FH. Efficacy and safety of dual blockade of the renin-angiotensin system: meta-analysis of randomised trials. BMJ 2013;346:f360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Zhao P, Xu P, Wan C, Wang Z. Evening versus morning dosing regimen drug therapy for hypertension. Cochrane Database Syst Rev 2011. (10):CD004184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Hermida RC, Ayala DE, Mojón A, Fernández JR. Influence of time of day of blood pressure-lowering treatment on cardiovascular risk in hypertensive patients with type 2 diabetes. Diabetes Care 2011;34:1270–1276 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Rahman M, Greene T, Phillips RA, et al. A trial of 2 strategies to reduce nocturnal blood pressure in Blacks with chronic kidney disease. Hypertension 2013;61:82–88 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Nilsson E, Gasparini A, Ärnlöv J, et al. Incidence and determinants of hyperkalemia and hypokalemia in a large healthcare system. Int J Cardiol 2017;245:277–284 [DOI] [PubMed] [Google Scholar]
  • 78. Bandak G, Sang Y, Gasparini A, et al. Hyperkalemia after initiating renin-angiotensin system blockade: the Stockholm Creatinine Measurements (SCREAM) project. J Am Heart Assoc 2017;6:e005428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Hughes-Austin JM, Rifkin DE, Beben T, et al. The relation of serum potassium concentration with cardiovascular events and mortality in community-living individuals. Clin J Am Soc Nephrol 2017;12:245–252 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. James MT, Grams ME, Woodward M, et al.; CKD Prognosis Consortium . A meta-analysis of the association of estimated GFR, albuminuria, diabetes mellitus, and hypertension with acute kidney injury. Am J Kidney Dis 2015;66:602–612 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Williams B, MacDonald TM, Morant S, et al.; British Hypertension Society’s PATHWAY Studies Group . Spironolactone versus placebo, bisoprolol, and doxazosin to determine the optimal treatment for drug-resistant hypertension (PATHWAY-2): a randomised, double-blind, crossover trial. Lancet 2015;386:2059–2068 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Sato A, Hayashi K, Naruse M, Saruta T. Effectiveness of aldosterone blockade in patients with diabetic nephropathy. Hypertension 2003;41:64–68 [DOI] [PubMed] [Google Scholar]
  • 83. Mehdi UF, Adams-Huet B, Raskin P, Vega GL, Toto RD. Addition of angiotensin receptor blockade or mineralocorticoid antagonism to maximal angiotensin-converting enzyme inhibition in diabetic nephropathy. J Am Soc Nephrol 2009;20:2641–2650 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Bakris GL, Agarwal R, Chan JC, et al.; Mineralocorticoid Receptor Antagonist Tolerability Study–Diabetic Nephropathy (ARTS-DN) Study Group . Effect of finerenone on albuminuria in patients with diabetic nephropathy: a randomized clinical trial. JAMA 2015;314:884–894 [DOI] [PubMed] [Google Scholar]
  • 85. Jensen MD, Ryan DH, Apovian CM, et al.; American College of Cardiology/American Heart Association Task Force on Practice Guidelines; Obesity Society . 2013 AHA/ACC/TOS guideline for the management of overweight and obesity in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and The Obesity Society. J Am Coll Cardiol 2014;63(25 Pt B):2985–3023 [DOI] [PubMed] [Google Scholar]
  • 86. Eckel RH, Jakicic JM, Ard JD, et al. 2013 AHA/ACC guideline on lifestyle management to reduce cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Circulation 2013;129:S76–S99 [DOI] [PubMed] [Google Scholar]
  • 87. Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/AHA guideline on the primary prevention of cardiovascular disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019;140:e596–e646 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Chasman DI, Posada D, Subrahmanyan L, Cook NR, Stanton VP Jr, Ridker PM. Pharmacogenetic study of statin therapy and cholesterol reduction. JAMA 2004;291:2821–2827 [DOI] [PubMed] [Google Scholar]
  • 89. Meek C, Wierzbicki AS, Jewkes C, et al. Daily and intermittent rosuvastatin 5 mg therapy in statin intolerant patients: an observational study. Curr Med Res Opin 2012;28:371–378 [DOI] [PubMed] [Google Scholar]
  • 90. Mihaylova B, Emberson J, Blackwell L, et al.; Cholesterol Treatment Trialists’ (CTT) Collaborators . The effects of lowering LDL cholesterol with statin therapy in people at low risk of vascular disease: meta-analysis of individual data from 27 randomised trials. Lancet 2012;380:581–590 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Baigent C, Keech A, Kearney PM, et al.; Cholesterol Treatment Trialists’ (CTT) Collaborators . Efficacy and safety of cholesterol-lowering treatment: prospective meta-analysis of data from 90,056 participants in 14 randomised trials of statins. Lancet 2005;366:1267–1278 [DOI] [PubMed] [Google Scholar]
  • 92. Pyŏrälä K, Pedersen TR, Kjekshus J, Faergeman O, Olsson AG, Thorgeirsson G. Cholesterol lowering with simvastatin improves prognosis of diabetic patients with coronary heart disease. A subgroup analysis of the Scandinavian Simvastatin Survival Study (4S). Diabetes Care 1997;20:614–620 [DOI] [PubMed] [Google Scholar]
  • 93. Collins R, Armitage J, Parish S, Sleigh P; Heart Protection Study Collaborative Group . MRC/BHF Heart Protection Study of cholesterol-lowering with simvastatin in 5963 people with diabetes: a randomised placebo-controlled trial. Lancet 2003;361:2005–2016 [DOI] [PubMed] [Google Scholar]
  • 94. Goldberg RB, Mellies MJ, Sacks FM, et al.; The Care Investigators . Cardiovascular events and their reduction with pravastatin in diabetic and glucose-intolerant myocardial infarction survivors with average cholesterol levels: subgroup analyses in the Cholesterol and Recurrent Events (CARE) trial. Circulation 1998;98:2513–2519 [DOI] [PubMed] [Google Scholar]
  • 95. Shepherd J, Barter P, Carmena R, et al. Effect of lowering LDL cholesterol substantially below currently recommended levels in patients with coronary heart disease and diabetes: the Treating to New Targets (TNT) study. Diabetes Care 2006;29:1220–1226 [DOI] [PubMed] [Google Scholar]
  • 96. Sever PS, Poulter NR, Dahlöf B, et al. Reduction in cardiovascular events with atorvastatin in 2,532 patients with type 2 diabetes: Anglo-Scandinavian Cardiac Outcomes Trial--lipid-lowering arm (ASCOT-LLA). Diabetes Care 2005;28:1151–1157 [DOI] [PubMed] [Google Scholar]
  • 97. Knopp RH, d’Emden M, Smilde JG, Pocock SJ. Efficacy and safety of atorvastatin in the prevention of cardiovascular end points in subjects with type 2 diabetes: the Atorvastatin Study for Prevention of Coronary Heart Disease Endpoints in non-insulin-dependent diabetes mellitus (ASPEN). Diabetes Care 2006;29:1478–1485 [DOI] [PubMed] [Google Scholar]
  • 98. Colhoun HM, Betteridge DJ, Durrington PN, et al.; CARDS investigators . Primary prevention of cardiovascular disease with atorvastatin in type 2 diabetes in the Collaborative Atorvastatin Diabetes Study (CARDS): multicentre randomised placebo-controlled trial. Lancet 2004;364:685–696 [DOI] [PubMed] [Google Scholar]
  • 99. Kearney PM, Blackwell L, Collins R, et al.; Cholesterol Treatment Trialists’ (CTT) Collaborators . Efficacy of cholesterol-lowering therapy in 18,686 people with diabetes in 14 randomised trials of statins: a meta-analysis. Lancet 2008;371:117–125 [DOI] [PubMed] [Google Scholar]
  • 100. Taylor F, Huffman MD, Macedo AF, et al. Statins for the primary prevention of cardiovascular disease. Cochrane Database Syst Rev 2013;1:CD004816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Carter AA, Gomes T, Camacho X, Juurlink DN, Shah BR, Mamdani MM. Risk of incident diabetes among patients treated with statins: population based study. BMJ 2013;346:f2610–f2610 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Jellinger PS, Handelsman Y, Rosenblit PD, et al. American Association of Clinical Endocrinologists and American College of Endocrinology guidelines for management of dyslipidemia and prevention of cardiovascular disease. Endocr Pract 2017;23(Suppl. 2):1–87 [DOI] [PubMed] [Google Scholar]
  • 103. Goldberg RB, Stone NJ, Grundy SM. The 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guidelines on the management of blood cholesterol in diabetes. Diabetes Care 2020;43:1673–1678 [DOI] [PubMed] [Google Scholar]
  • 104. Mach F, Baigent C, Catapano AL, et al.; ESC Scientific Document Group . 2019 ESC/EAS guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J 2020;41:111–188 [DOI] [PubMed] [Google Scholar]
  • 105. Cannon CP, Blazing MA, Giugliano RP, et al.; IMPROVE-IT Investigators . Ezetimibe added to statin therapy after acute coronary syndromes. N Engl J Med 2015;372:2387–2397 [DOI] [PubMed] [Google Scholar]
  • 106. de Ferranti SD, de Boer IH, Fonseca V, et al. Type 1 diabetes mellitus and cardiovascular disease: a scientific statement from the American Heart Association and American Diabetes Association. Diabetes Care 2014;37:2843–2863 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Cannon CP, Braunwald E, McCabe CH, et al.; Pravastatin or Atorvastatin Evaluation and Infection Therapy-Thrombolysis in Myocardial Infarction 22 Investigators . Intensive versus moderate lipid lowering with statins after acute coronary syndromes. N Engl J Med 2004;350:1495–1504 [DOI] [PubMed] [Google Scholar]
  • 108. Sabatine MS, Giugliano RP, Keech AC, et al.; FOURIER Steering Committee and Investigators . Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med 2017;376:1713–1722 [DOI] [PubMed] [Google Scholar]
  • 109. Giugliano RP, Cannon CP, Blazing MA, et al.; IMPROVE-IT (Improved Reduction of Outcomes: Vytorin Efficacy International Trial) Investigators . Benefit of adding ezetimibe to statin therapy on cardiovascular outcomes and safety in patients with versus without diabetes mellitus: results from IMPROVE-IT (Improved Reduction of Outcomes: Vytorin Efficacy International Trial). Circulation 2018;137:1571–1582 [DOI] [PubMed] [Google Scholar]
  • 110. Schwartz GG, Steg PG, Szarek M, et al.; ODYSSEY OUTCOMES Committees and Investigators . Alirocumab and cardiovascular outcomes after acute coronary syndrome. N Engl J Med 2018;379:2097–2107 [DOI] [PubMed] [Google Scholar]
  • 111. Ray KK, Colhoun HM, Szarek M, et al.; ODYSSEY OUTCOMES Committees and Investigators . Effects of alirocumab on cardiovascular and metabolic outcomes after acute coronary syndrome in patients with or without diabetes: a prespecified analysis of the ODYSSEY OUTCOMES randomised controlled trial. Lancet Diabetes Endocrinol 2019;7:618–628 [DOI] [PubMed] [Google Scholar]
  • 112. Sabatine MS, Leiter LA, Wiviott SD, et al. Cardiovascular safety and efficacy of the PCSK9 inhibitor evolocumab in patients with and without diabetes and the effect of evolocumab on glycaemia and risk of new-onset diabetes: a prespecified analysis of the FOURIER randomised controlled trial. Lancet Diabetes Endocrinol 2017;5:941–950 [DOI] [PubMed] [Google Scholar]
  • 113. Moriarty PM, Jacobson TA, Bruckert E, et al. Efficacy and safety of alirocumab, a monoclonal antibody to PCSK9, in statin-intolerant patients: design and rationale of ODYSSEY ALTERNATIVE, a randomized phase 3 trial. J Clin Lipidol 2014;8:554–561 [DOI] [PubMed] [Google Scholar]
  • 114. Zhang XL, Zhu QQ, Zhu L, et al. Safety and efficacy of anti-PCSK9 antibodies: a meta-analysis of 25 randomized, controlled trials. BMC Med 2015;13:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Giugliano RP, Pedersen TR, Saver JL, et al.; FOURIER Investigators . Stroke prevention with the PCSK9 (proprotein convertase subtilisin-kexin type 9) inhibitor evolocumab added to statin in high-risk patients with stable atherosclerosis. Stroke 2020;51:1546–1554 [DOI] [PubMed] [Google Scholar]
  • 116. Ray KK, Wright RS, Kallend D, et al.; ORION-10 and ORION-11 Investigators . Two phase 3 trials of inclisiran in patients with elevated LDL cholesterol. N Engl J Med 2020;382:1507–1519 [DOI] [PubMed] [Google Scholar]
  • 117. University of Oxford . A Randomized Trial Assessing the Effects of Inclisiran on Clinical Outcomes Among People With Cardiovascular Disease (ORION-4). In: ClinicalTrials.gov. Bethesda, MD, National Library of Medicine. NLM Identifier: NCT03705234. Accessed 15 October 2022. Available from https://clinicaltrials.gov/ct2/show/NCT03705234
  • 118. Dai L, Zuo Y, You Q, Zeng H, Cao S. Efficacy and safety of bempedoic acid in patients with hypercholesterolemia: a systematic review and meta-analysis of randomized controlled trials. Eur J Prev Cardiol 2021;28:825–83 [DOI] [PubMed] [Google Scholar]
  • 119. Di Minno A, Lupoli R, Calcaterra I, et al. Efficacy and safety of bempedoic acid in patients with hypercholesterolemia: systematic review and meta-analysis of randomized controlled trials. J Am Heart Assoc 2020;9:e016262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Berglund L, Brunzell JD, Goldberg AC, et al.; Endocrine society . Evaluation and treatment of hypertriglyceridemia: an Endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2012;97:2969–2989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121. Bhatt DL, Steg PG, Miller M, et al.; REDUCE-IT Investigators . Cardiovascular risk reduction with icosapent ethyl for hypertriglyceridemia. N Engl J Med 2019;380:11–22 [DOI] [PubMed] [Google Scholar]
  • 122. Nicholls SJ, Lincoff AM, Garcia M, et al. Effect of high-dose omega-3 fatty acids vs corn oil on major adverse cardiovascular events in patients at high cardiovascular risk: the STRENGTH randomized clinical trial. JAMA 2020;324:2268–2280 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Singh IM, Shishehbor MH, Ansell BJ. High-density lipoprotein as a therapeutic target: a systematic review. JAMA 2007;298:786–798 [DOI] [PubMed] [Google Scholar]
  • 124. Keech A, Simes RJ, Barter P, et al.; FIELD study investigators . Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet 2005;366:1849–1861 [DOI] [PubMed] [Google Scholar]
  • 125. Jones PH, Davidson MH. Reporting rate of rhabdomyolysis with fenofibrate + statin versus gemfibrozil + any statin. Am J Cardiol 2005;95:120–122 [DOI] [PubMed] [Google Scholar]
  • 126. Ginsberg HN, Elam MB, Lovato LC, et al.; ACCORD Study Group . Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med 2010;362:1563–1574 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Boden WE, Probstfield JL, Anderson T, et al.; AIM-HIGH Investigators . Niacin in patients with low HDL cholesterol levels receiving intensive statin therapy. N Engl J Med 2011;365:2255–2267 [DOI] [PubMed] [Google Scholar]
  • 128. Landray MJ, Haynes R, Hopewell JC, et al.; HPS2-THRIVE Collaborative Group . Effects of extended-release niacin with laropiprant in high-risk patients. N Engl J Med 2014;371:203–212 [DOI] [PubMed] [Google Scholar]
  • 129. Rajpathak SN, Kumbhani DJ, Crandall J, Barzilai N, Alderman M, Ridker PM. Statin therapy and risk of developing type 2 diabetes: a meta-analysis. Diabetes Care 2009;32:1924–1929 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Sattar N, Preiss D, Murray HM, et al. Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet 2010;375:735–742 [DOI] [PubMed] [Google Scholar]
  • 131. Ridker PM, Pradhan A, MacFadyen JG, Libby P, Glynn RJ. Cardiovascular benefits and diabetes risks of statin therapy in primary prevention: an analysis from the JUPITER trial. Lancet 2012;380:565–571 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Mach F, Ray KK, Wiklund O, et al.; European Atherosclerosis Society Consensus Panel . Adverse effects of statin therapy: perception vs. the evidence - focus on glucose homeostasis, cognitive, renal and hepatic function, haemorrhagic stroke and cataract. Eur Heart J 2018;39:2526–2539 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Heart Protection Study Collaborative Group . MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high-risk individuals: a randomised placebo-controlled trial. Lancet 2002;360:7–2212114036 [Google Scholar]
  • 134. Shepherd J, Blauw GJ, Murphy MB, et al.; PROSPER study group. PROspective Study of Pravastatin in the Elderly at Risk . Pravastatin in elderly individuals at risk of vascular disease (PROSPER): a randomised controlled trial. Lancet 2002;360:1623–1630 [DOI] [PubMed] [Google Scholar]
  • 135. Trompet S, van Vliet P, de Craen AJM, et al. Pravastatin and cognitive function in the elderly. Results of the PROSPER study. J Neurol 2010;257:85–90 [DOI] [PubMed] [Google Scholar]
  • 136. Yusuf S, Bosch J, Dagenais G, et al.; HOPE-3 Investigators . Cholesterol lowering in intermediate-risk persons without cardiovascular disease. N Engl J Med 2016;374:2021–2031 [DOI] [PubMed] [Google Scholar]
  • 137. Giugliano RP, Mach F, Zavitz K, et al.; EBBINGHAUS Investigators . Cognitive function in a randomized trial of evolocumab. N Engl J Med 2017;377:633–643 [DOI] [PubMed] [Google Scholar]
  • 138. Richardson K, Schoen M, French B, et al. Statins and cognitive function: a systematic review. Ann Intern Med 2013;159:688–697 [DOI] [PubMed] [Google Scholar]
  • 139. Baigent C, Blackwell L, Collins R, et al.; Antithrombotic Trialists’ (ATT) Collaboration . Aspirin in the primary and secondary prevention of vascular disease: collaborative meta-analysis of individual participant data from randomised trials. Lancet 2009;373:1849–1860 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Perk J, De Backer G, Gohlke H, et al.; European Association for Cardiovascular Prevention & Rehabilitation (EACPR); ESC Committee for Practice Guidelines (CPG) . European Guidelines on cardiovascular disease prevention in clinical practice (version 2012). The Fifth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of nine societies and by invited experts). Eur Heart J 2012;33:1635–1701 [DOI] [PubMed] [Google Scholar]
  • 141. Belch J, MacCuish A, Campbell I, et al.; Prevention of Progression of Arterial Disease and Diabetes Study Group; Diabetes Registry Group; Royal College of Physicians Edinburgh . The prevention of progression of arterial disease and diabetes (POPADAD) trial: factorial randomised placebo controlled trial of aspirin and antioxidants in patients with diabetes and asymptomatic peripheral arterial disease. BMJ 2008;337:a1840–a1840 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142. Zhang C, Sun A, Zhang P, et al. Aspirin for primary prevention of cardiovascular events in patients with diabetes: A meta-analysis. Diabetes Res Clin Pract 2010;87:211–218 [DOI] [PubMed] [Google Scholar]
  • 143. De Berardis G, Sacco M, Strippoli GFM, et al. Aspirin for primary prevention of cardiovascular events in people with diabetes: meta-analysis of randomised controlled trials. BMJ 2009;339:b4531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. ASCEND Study Collaborative Group . Effects of aspirin for primary prevention in persons with diabetes mellitus. N Engl J Med 2018;379:1529–1539 [DOI] [PubMed] [Google Scholar]
  • 145. Gaziano JM, Brotons C, Coppolecchia R, et al.; ARRIVE Executive Committee . Use of aspirin to reduce risk of initial vascular events in patients at moderate risk of cardiovascular disease (ARRIVE): a randomised, double-blind, placebo-controlled trial. Lancet 2018;392:1036–1046 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. McNeil JJ, Wolfe R, Woods RL, et al.; ASPREE Investigator Group . Effect of aspirin on cardiovascular events and bleeding in the healthy elderly. N Engl J Med 2018;379:1509–1518 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Pignone M, Earnshaw S, Tice JA, Pletcher MJ. Aspirin, statins, or both drugs for the primary prevention of coronary heart disease events in men: a cost-utility analysis. Ann Intern Med 2006;144:326–336 [DOI] [PubMed] [Google Scholar]
  • 148. Huxley RR, Peters SAE, Mishra GD, Woodward M. Risk of all-cause mortality and vascular events in women versus men with type 1 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol 2015;3:198–206 [DOI] [PubMed] [Google Scholar]
  • 149. Peters SAE, Huxley RR, Woodward M. Diabetes as risk factor for incident coronary heart disease in women compared with men: a systematic review and meta-analysis of 64 cohorts including 858,507 individuals and 28,203 coronary events. Diabetologia 2014;57:1542–1551 [DOI] [PubMed] [Google Scholar]
  • 150. Kalyani RR, Lazo M, Ouyang P, et al. Sex differences in diabetes and risk of incident coronary artery disease in healthy young and middle-aged adults. Diabetes Care 2014;37:830–838 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Peters SAE, Huxley RR, Woodward M. Diabetes as a risk factor for stroke in women compared with men: a systematic review and meta-analysis of 64 cohorts, including 775,385 individuals and 12,539 strokes. Lancet 2014;383:1973–1980 [DOI] [PubMed] [Google Scholar]
  • 152. Miedema MD, Duprez DA, Misialek JR, et al. Use of coronary artery calcium testing to guide aspirin utilization for primary prevention: estimates from the multi-ethnic study of atherosclerosis. Circ Cardiovasc Qual Outcomes 2014;7:453–460 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. Dimitriu-Leen AC, Scholte AJHA, van Rosendael AR, et al. Value of coronary computed tomography angiography in tailoring aspirin therapy for primary prevention of atherosclerotic events in patients at high risk with diabetes mellitus. Am J Cardiol 2016;117:887–893 [DOI] [PubMed] [Google Scholar]
  • 154. Mora S, Ames JM, Manson JE. Low-dose aspirin in the primary prevention of cardiovascular disease: shared decision making in clinical practice. JAMA 2016;316:709–710 [DOI] [PubMed] [Google Scholar]
  • 155. Campbell CL, Smyth S, Montalescot G, Steinhubl SR. Aspirin dose for the prevention of cardiovascular disease: a systematic review. JAMA 2007;297:2018–2024 [DOI] [PubMed] [Google Scholar]
  • 156. Jones WS, Mulder H, Wruck LM, et al.; ADAPTABLE Team . Comparative effectiveness of aspirin dosing in cardiovascular disease. N Engl J Med 2021;384:1981–1990 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Davì G, Patrono C. Platelet activation and atherothrombosis. N Engl J Med 2007;357:2482–2494 [DOI] [PubMed] [Google Scholar]
  • 158. Larsen SB, Grove EL, Neergaard-Petersen S, Würtz M, Hvas AM, Kristensen SD. Determinants of reduced antiplatelet effect of aspirin in patients with stable coronary artery disease. PLoS One 2015;10:e0126767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159. Zaccardi F, Rizzi A, Petrucci G, et al. In vivo platelet activation and aspirin responsiveness in type 1 diabetes. Diabetes 2016;65:503–509 [DOI] [PubMed] [Google Scholar]
  • 160. Bethel MA, Harrison P, Sourij H, et al. Randomized controlled trial comparing impact on platelet reactivity of twice-daily with once-daily aspirin in people with type 2 diabetes. Diabet Med 2016;33:224–230 [DOI] [PubMed] [Google Scholar]
  • 161. Rothwell PM, Cook NR, Gaziano JM, et al. Effects of aspirin on risks of vascular events and cancer according to bodyweight and dose: analysis of individual patient data from randomised trials. Lancet 2018;392:387–399 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Vandvik PO, Lincoff AM, Gore JM, et al. Primary and secondary prevention of cardiovascular disease: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest 2012;141(Suppl.):e637S–e668S [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Bhatt DL, Bonaca MP, Bansilal S, et al. Reduction in ischemic events with ticagrelor in diabetic patients with prior myocardial infarction in PEGASUS-TIMI 54. J Am Coll Cardiol 2016;67:2732–2740 [DOI] [PubMed] [Google Scholar]
  • 164. Steg PG, Bhatt DL, Simon T, et al.; THEMIS Steering Committee and Investigators . Ticagrelor in patients with stable coronary disease and diabetes. N Engl J Med 2019;381:1309–1320 [DOI] [PubMed] [Google Scholar]
  • 165. Bhatt DL, Steg PG, Mehta SR, et al.; THEMIS Steering Committee and Investigators . Ticagrelor in patients with diabetes and stable coronary artery disease with a history of previous percutaneous coronary intervention (THEMIS-PCI): a phase 3, placebo-controlled, randomised trial. Lancet 2019;394:1169–1180 [DOI] [PubMed] [Google Scholar]
  • 166. Angiolillo DJ, Baber U, Sartori S, et al. Ticagrelor with or without aspirin in high-risk patients with diabetes mellitus undergoing percutaneous coronary intervention. J Am Coll Cardiol 2020;75:2403–2413 [DOI] [PubMed] [Google Scholar]
  • 167. Wiebe J, Ndrepepa G, Kufner S, et al. Early aspirin discontinuation after coronary stenting: a systematic review and meta-analysis. J Am Heart Assoc 2021;10:e018304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Bhatt DL, Eikelboom JW, Connolly SJ, et al.; COMPASS Steering Committee and Investigators . Role of combination antiplatelet and anticoagulation therapy in diabetes mellitus and cardiovascular disease: insights from the COMPASS trial. Circulation 2020;141:1841–1854 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. Connolly SJ, Eikelboom JW, Bosch J, et al.; COMPASS investigators . Rivaroxaban with or without aspirin in patients with stable coronary artery disease: an international, randomised, double-blind, placebo-controlled trial. Lancet 2018;391:205–218 [DOI] [PubMed] [Google Scholar]
  • 170. Bonaca MP, Bauersachs RM, Anand SS, et al. Rivaroxaban in peripheral artery disease after revascularization. N Engl J Med 2020;382:1994–2004 [DOI] [PubMed] [Google Scholar]
  • 171. Bax JJ, Young LH, Frye RL, Bonow RO, Steinberg HO; ADA . Screening for coronary artery disease in patients with diabetes. Diabetes Care 2007;30:2729–2736 [DOI] [PubMed] [Google Scholar]
  • 172. Boden WE, O’Rourke RA, Teo KK, et al.; COURAGE Trial Research Group . Optimal medical therapy with or without PCI for stable coronary disease. N Engl J Med 2007;356:1503–1516 [DOI] [PubMed] [Google Scholar]
  • 173. Frye RL, August P, Brooks MM, et al.; BARI 2D Study Group . A randomized trial of therapies for type 2 diabetes and coronary artery disease. N Engl J Med 2009;360:2503–2515 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Wackers FJT, Chyun DA, Young LH, et al.; Detection of Ischemia in Asymptomatic Diabetics (DIAD) Investigators . Resolution of asymptomatic myocardial ischemia in patients with type 2 diabetes in the Detection of Ischemia in Asymptomatic Diabetics (DIAD) study. Diabetes Care 2007;30:2892–2898 [DOI] [PubMed] [Google Scholar]
  • 175. Elkeles RS, Godsland IF, Feher MD, et al.; PREDICT Study Group . Coronary calcium measurement improves prediction of cardiovascular events in asymptomatic patients with type 2 diabetes: the PREDICT study. Eur Heart J 2008;29:2244–2251 [DOI] [PubMed] [Google Scholar]
  • 176. Raggi P, Shaw LJ, Berman DS, Callister TQ. Prognostic value of coronary artery calcium screening in subjects with and without diabetes. J Am Coll Cardiol 2004;43:1663–1669 [DOI] [PubMed] [Google Scholar]
  • 177. Anand DV, Lim E, Hopkins D, et al. Risk stratification in uncomplicated type 2 diabetes: prospective evaluation of the combined use of coronary artery calcium imaging and selective myocardial perfusion scintigraphy. Eur Heart J 2006;27:713–721 [DOI] [PubMed] [Google Scholar]
  • 178. Young LH, Wackers FJT, Chyun DA, et al.; DIAD Investigators . Cardiac outcomes after screening for asymptomatic coronary artery disease in patients with type 2 diabetes: the DIAD study: a randomized controlled trial. JAMA 2009;301:1547–1555 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Wackers FJT, Young LH, Inzucchi SE, et al.; Detection of Ischemia in Asymptomatic Diabetics Investigators . Detection of silent myocardial ischemia in asymptomatic diabetic subjects: the DIAD study. Diabetes Care 2004;27:1954–1961 [DOI] [PubMed] [Google Scholar]
  • 180. Scognamiglio R, Negut C, Ramondo A, Tiengo A, Avogaro A. Detection of coronary artery disease in asymptomatic patients with type 2 diabetes mellitus. J Am Coll Cardiol 2006;47:65–71 [DOI] [PubMed] [Google Scholar]
  • 181. Hadamitzky M, Hein F, Meyer T, et al. Prognostic value of coronary computed tomographic angiography in diabetic patients without known coronary artery disease. Diabetes Care 2010;33:1358–1363 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182. Choi EK, Chun EJ, Choi SI, et al. Assessment of subclinical coronary atherosclerosis in asymptomatic patients with type 2 diabetes mellitus with single photon emission computed tomography and coronary computed tomography angiography. Am J Cardiol 2009;104:890–896 [DOI] [PubMed] [Google Scholar]
  • 183. Malik S, Zhao Y, Budoff M, et al. Coronary artery calcium score for long-term risk classification in individuals with type 2 diabetes and metabolic syndrome from the Multi-Ethnic Study of Atherosclerosis. JAMA Cardiol 2017;2:1332–1340 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184. Wing RR, Bolin P, Brancati FL, et al.; Look AHEAD Research Group . Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. N Engl J Med 2013;369:145–154 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Braunwald E, Domanski MJ, Fowler SE, et al.; PEACE Trial Investigators . Angiotensin-converting-enzyme inhibition in stable coronary artery disease. N Engl J Med 2004;351:2058–2068 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186. Pitt B, Filippatos G, Agarwal R, et al.; FIGARO-DKD Investigators . Cardiovascular events with finerenone in kidney disease and type 2 diabetes. N Engl J Med 2021;385:2252–2263 [DOI] [PubMed] [Google Scholar]
  • 187. Filippatos G, Anker SD, Agarwal R, et al.; FIGARO-DKD Investigators . Finerenone reduces risk of incident heart failure in patients with chronic kidney disease and type 2 diabetes: analyses from the FIGARO-DKD trial. Circulation 2022;145:437–447 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188. Agarwal R, Filippatos G, Pitt B, et al.; FIDELIO-DKD and FIGARO-DKD investigators . Cardiovascular and kidney outcomes with finerenone in patients with type 2 diabetes and chronic kidney disease: the FIDELITY pooled analysis. Eur Heart J 2022;43:474–484 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189. Anker SD, Butler J, Filippatos G, et al.; EMPEROR-Preserved Trial Investigators . Empagliflozin in heart failure with a preserved ejection fraction. N Engl J Med 2021;385:1451–1461 [DOI] [PubMed] [Google Scholar]
  • 190. Kezerashvili A, Marzo K, De Leon J. Beta blocker use after acute myocardial infarction in the patient with normal systolic function: when is it “ok” to discontinue? Curr Cardiol Rev 2012;8:77–84 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191. Fihn SD, Gardin JM, Abrams J, et al.; American College of Cardiology Foundation; American Heart Association Task Force on Practice Guidelines; American College of Physicians; American Association for Thoracic Surgery; Preventive Cardiovascular Nurses Association; Society for Cardiovascular Angiography and Interventions; Society of Thoracic Surgeons . 2012 ACCF/AHA/ACP/AATS/PCNA/SCAI/STS Guideline for the diagnosis and management of patients with stable ischemic heart disease: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines, and the American College of Physicians, American Association for Thoracic Surgery, Preventive Cardiovascular Nurses Association, Society for Cardiovascular Angiography and Interventions, and Society of Thoracic Surgeons. J Am Coll Cardiol 2012;60:e44–e164 [DOI] [PubMed] [Google Scholar]
  • 192. U.S. Food and Drug Administration . Guidance for industry. Diabetes mellitus—evaluating cardiovascular risk in new antidiabetic therapies to treat type 2 diabetes. Silver Spring, MD, 2008. Accessed 21 October 2022. Available from https://www.federalregister.gov/documents/2008/12/19/E8-30086/guidance-for-industry-on-diabetes-mellitus-evaluating-cardiovascular-risk-in-new-antidiabetic
  • 193. Rosenstock J, Perkovic V, Johansen OE, et al.; CARMELINA Investigators . Effect of linagliptin vs placebo on major cardiovascular events in adults with type 2 diabetes and high cardiovascular and renal risk: the CARMELINA randomized clinical trial. JAMA 2019;321:69–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194. Perkovic V, Jardine MJ, Neal B, et al.; CREDENCE Trial Investigators . Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med 2019;380:2295–2306 [DOI] [PubMed] [Google Scholar]
  • 195. Neal B, Perkovic V, Matthews DR, et al.; CANVAS-R Trial Collaborative Group . Rationale, design and baseline characteristics of the CANagliflozin cardioVascular Assessment Study-Renal (CANVAS-R): A randomized, placebo-controlled trial. Diabetes Obes Metab 2017;19:387–393 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Wiviott SD, Raz I, Bonaca MP, et al.; DECLARE–TIMI 58 Investigators . Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2019;380:347–357 [DOI] [PubMed] [Google Scholar]
  • 197. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al.; DAPA-CKD Trial Committees and Investigators . Dapagliflozin in patients with chronic kidney disease. N Engl J Med 2020;383:1436–1446 [DOI] [PubMed] [Google Scholar]
  • 198. Nassif ME, Windsor SL, Borlaug BA, et al. The SGLT2 inhibitor dapagliflozin in heart failure with preserved ejection fraction: a multicenter randomized trial. Nat Med 2021;27:1954–1960 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Solomon SD, McMurray JJV, Claggett B, et al.; DELIVER Trial Committees and Investigators . Dapagliflozin in heart failure with mildly reduced or preserved ejection fraction. N Engl J Med 2022;387:1089–1098 [DOI] [PubMed] [Google Scholar]
  • 200. Packer M, Anker SD, Butler J, et al.; EMPEROR Reduced Trial Investigators . Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med 2020;383:1413–1424 [DOI] [PubMed] [Google Scholar]
  • 201. Cannon CP, Pratley R, Dagogo-Jack S, et al.; VERTIS CV Investigators . Cardiovascular outcomes with ertugliflozin in type 2 diabetes. N Engl J Med 2020;383:1425–1435 [DOI] [PubMed] [Google Scholar]
  • 202. Bhatt DL, Szarek M, Pitt B, et al.; SCORED Investigators . Sotagliflozin in patients with diabetes and chronic kidney disease. N Engl J Med 2021;384:129–139 [DOI] [PubMed] [Google Scholar]
  • 203. Marso SP, Daniels GH, Brown-Frandsen K, et al.; LEADER Steering Committee; LEADER Trial Investigators . Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2016;375:311–322 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204. Marso SP, Bain SC, Consoli A, et al.; SUSTAIN-6 Investigators . Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2016;375:1834–1844 [DOI] [PubMed] [Google Scholar]
  • 205. Husain M, Birkenfeld AL, Donsmark M, et al. Oral semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2019;381:841–851 [DOI] [PubMed] [Google Scholar]
  • 206. Hernandez AF, Green JB, Janmohamed S, et al.; Harmony Outcomes committees and investigators . Albiglutide and cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease (Harmony Outcomes): a double-blind, randomised placebo-controlled trial. Lancet 2018;392:1519–1529 [DOI] [PubMed] [Google Scholar]
  • 207. Gerstein HC, Colhoun HM, Dagenais GR, et al.; REWIND Investigators . Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 2019;394:121–130 [DOI] [PubMed] [Google Scholar]
  • 208. Pfeffer MA, Claggett B, Diaz R, et al.; ELIXA Investigators . Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med 2015;373:2247–2257 [DOI] [PubMed] [Google Scholar]
  • 209. Holman RR, Bethel MA, Mentz RJ, et al.; EXSCEL Study Group . Effects of once-weekly exenatide on cardiovascular outcomes in type 2 diabetes. N Engl J Med 2017;377:1228–1239 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210. Zelniker TA, Wiviott SD, Raz I, et al. Comparison of the effects of glucagon-like peptide receptor agonists and sodium-glucose cotransporter 2 inhibitors for prevention of major adverse cardiovascular and renal outcomes in type 2 diabetes mellitus. Circulation 2019;139:2022–2031 [DOI] [PubMed] [Google Scholar]
  • 211. Palmer SC, Tendal B, Mustafa RA, et al. Sodium-glucose cotransporter protein-2 (SGLT-2) inhibitors and glucagon-like peptide-1 (GLP-1) receptor agonists for type 2 diabetes: systematic review and network meta-analysis of randomised controlled trials. BMJ 2021;372:m4573. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212. Zelniker TA, Wiviott SD, Raz I, et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet 2019;393:31–39 [DOI] [PubMed] [Google Scholar]
  • 213. McGuire DK, Shih WJ, Cosentino F, et al. Association of SGLT2 inhibitors with cardiovascular and kidney outcomes in patients with type 2 diabetes: a meta-analysis. JAMA Cardiol 2021;6:148–158 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214. Gerstein HC, Sattar N, Rosenstock J, et al. Cardiovascular and renal outcomes with efpeglenatide in type 2 diabetes. N Engl J Med 2021;385:896–907 [DOI] [PubMed] [Google Scholar]
  • 215. Lam CSP, Ramasundarahettige C, Branch KRH, et al. Efpeglenatide and clinical outcomes with and without concomitant sodium-glucose cotransporter-2 inhibition use in type 2 diabetes: exploratory analysis of the AMPLITUDE-O Trial. Circulation 2022;145:565–574 [DOI] [PubMed] [Google Scholar]
  • 216. Kannel WB, Hjortland M, Castelli WP. Role of diabetes in congestive heart failure: the Framingham study. Am J Cardiol 1974;34:29–34 [DOI] [PubMed] [Google Scholar]
  • 217. Dunlay SM, Givertz MM, Aguilar D, et al.; American Heart Association Heart Failure and Transplantation Committee of the Council on Clinical Cardiology; Council on Cardiovascular and Stroke Nursing; Heart Failure Society of America . Type 2 diabetes mellitus and heart failure, a scientific statement from the American Heart Association and Heart Failure Society of America. J Card Fail 2019;25:584–619 [DOI] [PubMed] [Google Scholar]
  • 218. Dormandy JA, Charbonnel B, Eckland DJA, et al.; PROactive Investigators . Secondary prevention of macrovascular events in patients with type 2 diabetes in the PROactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial. Lancet 2005;366:1279–1289 [DOI] [PubMed] [Google Scholar]
  • 219. Singh S, Loke YK, Furberg CD. Long-term risk of cardiovascular events with rosiglitazone: a meta-analysis. JAMA 2007;298:1189–1195 [DOI] [PubMed] [Google Scholar]
  • 220. Lincoff AM, Wolski K, Nicholls SJ, Nissen SE. Pioglitazone and risk of cardiovascular events in patients with type 2 diabetes mellitus: a meta-analysis of randomized trials. JAMA 2007;298:1180–1188 [DOI] [PubMed] [Google Scholar]
  • 221. Inzucchi SE, Masoudi FA, McGuire DK. Metformin in heart failure. Diabetes Care 2007;30:e129–e129 [DOI] [PubMed] [Google Scholar]
  • 222. Eurich DT, Majumdar SR, McAlister FA, Tsuyuki RT, Johnson JA. Improved clinical outcomes associated with metformin in patients with diabetes and heart failure. Diabetes Care 2005;28:2345–2351 [DOI] [PubMed] [Google Scholar]
  • 223. 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, 2016. Accessed 21 October 2022. Available from https://www.fda.gov/drugs/drug-safety-and-availability/fda-drug-safety-communication-fda-revises-warnings-regarding-use-diabetes-medicine-metformin-certain
  • 224. Scirica BM, Bhatt DL, Braunwald E, et al.; SAVOR-TIMI 53 Steering Committee and Investigators . Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med 2013;369:1317–1326 [DOI] [PubMed] [Google Scholar]
  • 225. Zannad F, Cannon CP, Cushman WC, et al.; EXAMINE Investigators . Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a multicentre, randomised, double-blind trial. Lancet 2015;385:2067–2076 [DOI] [PubMed] [Google Scholar]
  • 226. Green JB, Bethel MA, Armstrong PW, et al.; TECOS Study Group . Effect of sitagliptin on cardiovascular outcomes in type 2 diabetes. N Engl J Med 2015;373:232–242 [DOI] [PubMed] [Google Scholar]
  • 227. Vaduganathan M, Docherty KF, Claggett BL, et al. SGLT-2 inhibitors in patients with heart failure: a comprehensive meta-analysis of five randomised controlled trials. Lancet 2022;400:757–767 [DOI] [PubMed] [Google Scholar]
  • 228. Bhatt DL, Szarek M, Steg PG, et al.; SOLOIST-WHF Trial Investigators . Sotagliflozin in patients with diabetes and recent worsening heart failure. N Engl J Med 2021;384:117–128 [DOI] [PubMed] [Google Scholar]
  • 229. Packer M, Anker SD, Butler J, et al. Effect of empagliflozin on the clinical stability of patients with heart failure and a reduced ejection fraction: the EMPEROR-Reduced trial. Circulation 2021;143:326–336 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230. Voors AA, Angermann CE, Teerlink JR, et al. The SGLT2 inhibitor empagliflozin in patients hospitalized for acute heart failure: a multinational randomized trial. Nat Med 2022;28:568–574 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231. Spertus JA, Birmingham MC, Nassif M, et al. The SGLT2 inhibitor canagliflozin in heart failure: the CHIEF-HF remote, patient-centered randomized trial. Nat Med 2022;28:809–813 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232. Gansevoort RT, Correa-Rotter R, Hemmelgarn BR, et al. Chronic kidney disease and cardiovascular risk: epidemiology, mechanisms, and prevention. Lancet 2013;382:339–352 [DOI] [PubMed] [Google Scholar]
  • 233. Bakris GL, Agarwal R, Anker SD, et al.; FIDELIO-DKD Investigators . Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. N Engl J Med 2020;383:2219–2229 [DOI] [PubMed] [Google Scholar]
  • 234. Das SR, Everett BM, Birtcher KK, et al. 2020 expert consensus decision pathway on novel therapies for cardiovascular risk reduction in patients with type 2 diabetes: a report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol 2020;76:1117–1145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235. White WB, Cannon CP, Heller SR, et al.; EXAMINE Investigators . Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N Engl J Med 2013;369:1327–1335 [DOI] [PubMed] [Google Scholar]
  • 236. Rosenstock J, Perkovic V, Alexander JH, et al.; CARMELINA investigators . Rationale, design, and baseline characteristics of the CArdiovascular safety and Renal Microvascular outcomE study with LINAgliptin (CARMELINA): a randomized, double-blind, placebo-controlled clinical trial in patients with type 2 diabetes and high cardio-renal risk. Cardiovasc Diabetol 2018;17:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237. Marx N, Rosenstock J, Kahn SE, et al. Design and baseline characteristics of the CARdiovascular Outcome Trial of LINAgliptin Versus Glimepiride in Type 2 Diabetes (CAROLINA®). Diab Vasc Dis Res 2015;12:164–174 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238. Cefalu WT, Kaul S, Gerstein HC, et al. Cardiovascular outcomes trials in type 2 diabetes: where do we go from here? Reflections from a Diabetes Care Editors’ Expert Forum. Diabetes Care 2018;41:14–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239. Wheeler DC, Stefansson BV, Batiushin M, et al. The dapagliflozin and prevention of adverse outcomes in chronic kidney disease (DAPA-CKD) trial: baseline characteristics. Nephrol Dial Transplant 2020;35:1700–1711 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240. Cannon CP, McGuire DK, Pratley R, et al.; VERTIS-CV Investigators . Design and baseline characteristics of the eValuation of ERTugliflozin effIcacy and Safety CardioVascular outcomes trial (VERTIS-CV). Am Heart J 2018;206:11–23 [DOI] [PubMed] [Google Scholar]
  • 241. Anker SD, Butler J, Filippatos G, et al.; EMPEROR-Preserved Trial Committees and Investigators . Baseline characteristics of patients with heart failure with preserved ejection fraction in the EMPEROR-Preserved trial. Eur J Heart Fail 2020;22:2383–2392 [DOI] [PubMed] [Google Scholar]
  • 242. Zoungas S, Chalmers J, Neal B, et al.; ADVANCE-ON Collaborative Group . Follow-up of blood-pressure lowering and glucose control in type 2 diabetes. N Engl J Med 2014;371:1392–1406 [DOI] [PubMed] [Google Scholar]

Articles from Diabetes Care are provided here courtesy of American Diabetes Association

RESOURCES