U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

Lawrence JM, Casagrande SS, Herman WH, et al., editors. Diabetes in America [Internet]. Bethesda (MD): National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK); 2023-.

Cover of Diabetes in America

Diabetes in America [Internet].

Show details

Heart Disease and Diabetes

, MD, MHS, , MD, MPH, , MD, and , MD, MHS.

Author Information and Affiliations

Initial Posting: .

Summary

Heart disease remains a major cause of morbidity and mortality among individuals with type 2 diabetes. Meta-analyses have demonstrated a pooled relative risk for incident coronary heart disease (CHD) that is approximately twofold higher overall in adults with diabetes compared to those without diabetes. In studies that further stratify results by sex, the relative risk of CHD is higher in women than men in the presence of diabetes. Nonetheless, the age-standardized prevalence for all categories of heart disease remains higher in men with diabetes than women with diabetes in the United States. Although the rates of diabetes are higher in Hispanic and non-White individuals compared to non-Hispanic White adults, it should be noted that non-Hispanic White adults with diabetes generally report heart disease rates up to twice as high as Hispanic persons with diabetes, with an intermediate prevalence of any heart condition in non-Hispanic Black individuals. Despite a more than twofold increase in type 2 diabetes prevalence from the 1970s to the 2020s, the prevalence of heart disease has increased modestly in both men with diabetes and women with diabetes, while in contrast, it has remained stable or decreased for men and women without diabetes.

Classic heart disease risk markers have been clearly demonstrated to be important determinants of heart disease in diabetes, including elevated low-density lipoprotein cholesterol, elevated blood pressure, smoking, and elevated triglycerides and low high-density lipoprotein cholesterol. Obesity is an important risk factor for type 2 diabetes but has not consistently been shown to have an independent association with heart disease, possibly because obesity is in the causal pathway between these risk factors and heart disease development. However, several studies indicate that the excess prevalence of heart disease in diabetes is not fully accounted for by measured classic cardiovascular disease risk factors. In addition, novel biomarkers have been found to either add no or only modest incremental significance in the prediction of heart disease. The association between fasting glucose and heart disease displays a J-shaped curve in several studies. Glycosylated hemoglobin also has a graded association with heart disease. The association between insulin resistance and heart disease is inconsistent, at least in part because of methodologic differences among studies. Other important risk factors include lifestyle factors, such as physical activity, smoking, diet, and social determinants of health, such as food insecurity, access to health care, or poverty.

Clinical trials involving modification of cardiovascular risk factors in diabetes have helped to clarify their roles in heart disease development. Clinical trials focusing on weight reduction through an intensive lifestyle intervention specifically in people with diabetes have not demonstrated benefit in cardiovascular events despite improvement in risk factors. Whether improvement of glycemic control reduces heart disease has long been a central question, since older trials had not consistently demonstrated benefit. By contrast, lipid-lowering clinical trials have shown that statin treatment, in both secondary as well as primary prevention trials, significantly reduces atherosclerotic heart disease with a similar risk reduction to that seen in people without diabetes. Large randomized trials have demonstrated that highly purified eicosapentaenoic acid ethyl ester significantly reduces risk of ischemic events. Trials of more intensive compared to standard blood pressure control in people with diabetes did not generally find that achieving a lower goal leads to a reduction in cardiovascular events overall. The benefits of aspirin in reducing serious vascular events have been demonstrated in trials of people with diabetes, but these benefits are largely counterbalanced by an increase in major bleeding events.

Post-marketing cardiovascular outcome trials have been required by the U.S. Food and Drug Administration to demonstrate safety for all antihyperglycemic agents newly approved since 2008. All agents in the glucagon-like peptide-1 receptor agonist and sodium-glucose cotransporter-2 inhibitor classes have demonstrated safety, while some agents within these classes of medication have further demonstrated superiority in reducing major adverse cardiovascular events, hospitalization for heart failure, and kidney failure in specific populations.

In conclusion, despite intensive management of cardiovascular risk factors, the high risk for heart disease among people with diabetes remains a major health concern. Importantly, over the past few years, an increasing number of therapies have become available to reduce cardiovascular risk in people with type 2 diabetes.

Introduction

Although coronary heart disease (CHD) is the major cause of morbidity and mortality in patients with type 2 diabetes, historically the diabetes-CHD association received little systematic study, even after the publication of Kelly West’s monumental book Epidemiology of Diabetes Mellitus and Its Vascular Lesions (1) and the development of standard World Health Organization (WHO) (2) and National Diabetes Data Group (3) criteria for the definition of diabetes in the 1980s.

According to the Centers for Disease Control and Prevention (CDC) (4), the proportion of diabetes in the United States that is type 1 is 5%–10%, while type 2 diabetes accounts for 90%–95%. The epidemiology and etiology of type 1 (insulin-dependent) diabetes are described elsewhere (5). Aside from the Diabetes Control and Complications Trial (DCCT), there is a relative paucity of trials with cardiovascular outcomes in people with type 1 diabetes. Although longstanding type 1 diabetes carries a cardiovascular disease (CVD) mortality risk similar to type 2 diabetes (6), type 2 diabetes is the primary focus of the analyses herein, based on papers reporting diabetes defined by elevated fasting plasma glucose (FPG) values or diabetes by history.

The literature on diabetes and CVD, particularly from studies reporting the results from cardiovascular outcome trials (CVOTs) of newer glucose-lowering agents, has increased exponentially since Diabetes in America, 3rd edition, was published in 2018 (7,8). Here, each section usually starts with a review of the evidence and, whenever possible, concludes showing a large systematic review or meta-analysis that addresses each topic. In order to improve generalizability and statistical power, no attempt was made to exclude prospective studies done outside the United States that were included in these meta-analyses.

Diabetes is clearly an established risk factor for CHD (9,10), but how much its effect varies by age, sex, country, or levels of conventional risk factors for CHD remains unresolved (11). Key characteristics of diabetes are discussed, specifically: demographic and lifestyle factors; social determinants of health, such as food insecurity or access to health care; and metabolic risk factors, including hyperglycemia and insulin (hyperinsulinemia), dyslipidemia (elevated triglycerides, high low-density lipoprotein cholesterol [LDL-C], and low high-density lipoprotein cholesterol [HDL-C]), hypertension, obesity and components of the metabolic syndrome, and novel risk markers, such as high-sensitivity C-reactive protein (hsCRP). Hypertension is a common risk factor for CHD in adults with diabetes and is an independent risk factor for heart disease, heart failure (HF), and stroke. The extent to which diabetes is associated with fatal versus nonfatal myocardial infarction (MI) is also unknown (12). Further, how much of the effect of diabetes on vascular risk can be accounted for by conventional vascular risk factors (dyslipidemia, hypertension, obesity, smoking) is unresolved (13). FPG has been log-linearly and importantly associated with risk of vascular disease at all concentrations, including below the classic fasting threshold for diabetes of 126 mg/dL (6.99 mmol/L); however, available data regarding this association remain inconclusive (14,15,16).

Figure 1 shows study-specific hazard ratios (HR) for CHD in people with diabetes at baseline compared with people without diabetes (17). The summary risk estimates from the forest plot indicate that the overall effect is an approximate doubling of risk for CHD among those with diabetes compared to those without diabetes. Regression analyses were stratified, where appropriate, by sex and trial group and adjusted for age, smoking status, body mass index (BMI), and systolic blood pressure. Studies are ordered (top to bottom) by increasing number of CHD cases. Sizes of data markers are proportional to the inverse of the variance of the hazard ratios. The data sources shown in this forest plot reveal the heterogeneity of associations by cohort.

Forest plot showing that the risk of coronary heart disease is approximately double for those with diabetes compared to those without diabetes.

FIGURE 1.

Study-Specific Hazard Ratios for Coronary Heart Disease in People With Diabetes at Baseline Compared to People Without Diabetes. Regression analyses were stratified, where appropriate, by sex and trial group, and adjusted for age, smoking status, body (more...)

Obesity is increasing steadily worldwide, including in industrializing countries. In the United States, obesity prevalence in adults increased from 30.5% in 1999–2000 to 42.4% in 2017–2018 (18), yet the prevalence of obesity in persons with type 2 diabetes is even higher. According to the CDC and based on National Health and Nutrition Examination Survey (NHANES) data between 2015 and 2018, approximately 62% of adults with diabetes are obese and an additional 27.7% are overweight (19). While large research studies have demonstrated the effectiveness of lifestyle changes (dietary changes and increased physical activity) in reducing the incidence of diabetes, losing weight is difficult in real-world settings. Sustained weight loss, such as after bariatric surgery, may be required to produce meaningful changes in health outcomes, particularly for CVD.

In this article, the clinical trial evidence is reviewed for interventions designed to produce healthy changes in heart disease risk factors and reduce heart disease, comparing adults with or without diabetes. In particular, the results from CVOTs of newer glucose-lowering agents and implications for clinical care of people with diabetes are discussed.

Sources and Limitations of National Data on Heart Disease and Diabetes

Information on heart disease and diabetes is available from several surveys conducted in the United States that use national probability samples, including the National Health Interview Survey (NHIS), the NHANES, and the Behavioral Risk Factor Surveillance System (BRFSS). The NHIS is a cross-sectional household interview survey that uses a complex sampling design and has been conducted continuously since 1957. In the NHIS 2019–2020 survey cycles, participants were asked detailed questions about diabetes and heart disease. Diabetes was determined if participants answered “yes” to the following question: “(If female, other than during pregnancy) Have you ever been told by a doctor or health professional that you have diabetes or sugar diabetes?” Questions about heart disease included any history of congestive heart failure, coronary heart disease, angina, or heart attack. Although the heart disease questions in NHIS are comprehensive, a limitation is that the data are self-reported.

The NHANES is a cross-sectional, national probability sample that has been conducted periodically since 1971 and continuously since 1999. NHANES data collected from individuals for the period 2017–2020 Q1 are reported to assess current estimates in heart disease among adults with diabetes before the start of the coronavirus disease of 2019 (COVID-19) pandemic. In addition, data from survey periods between 1976–1980 and 2017–2020 Q1 are utilized to assess trends in heart disease. Participants self-reported diagnosed diabetes status, based on the question of “Other than during pregnancy, have you ever been told by a doctor or health professional that you have diabetes or sugar diabetes?” An advantage of the NHANES is that the survey includes a health exam in a mobile examination center where laboratory measures are collected. Thus, the NHANES has laboratory information on glycosylated hemoglobin (A1c) and FPG to determine undiagnosed diabetes. Participants self-reported any history of congestive heart failure, coronary heart disease, angina, or heart attack.

The BRFSS is a large, state-based, telephone-based survey conducted annually to collect data on respondents’ health-related behaviors, chronic health conditions, and use of preventive services. Data from 2019 are utilized to describe self-reported information on diagnosed diabetes, coronary heart disease or angina, and heart attack or MI.

The Healthcare Cost & Utilization Project (HCUP), National (Nationwide) Inpatient Sample (NIS) is the largest publicly available all-payer inpatient healthcare database designed to produce U.S. national estimates of in-patient utilization, access, cost, quality, and outcomes. The HCUP-NIS estimates around 35 million hospitalizations nationally. Data from the HCUP-NIS 2018 are utilized to estimate heart disease by diabetes status using ICD-10 (International Classification of Diseases, Tenth Revision) codes.

Statistical Methods

All estimates from national surveys are weighted to produce estimates that are nationally representative of the noninstitutionalized U.S. population. Weighted standard errors (SE) are provided for estimates in the tables. The relative standard error (RSE = [SE/estimate]*100) is provided in tables and figures for estimates that are likely unreliable due to sample size. Estimates with RSEs >50% are censored.

Prevalence of Heart Disease in People With Versus Without Diabetes in the United States: Nationally Representative Survey Estimates

National surveys of the U.S. population consistently demonstrate that the prevalence of heart disease (no matter how defined) is higher among adults with diabetes than adults without diabetes (Tables 1, 2, 3, and 4; Appendices A1, A2, A3, and A4). In adults, the prevalence of heart disease increases with age regardless of diabetes status (Figure 2). The age-standardized relative risk (RR; the ratio in persons with diabetes to those without) for any heart condition, including CHD, angina, and heart attack (plus HF in the NHANES), varies from 2.2 to 2.5, reflecting the different heart disease conditions included in the national surveys. Prevalence estimates for any heart condition are 20.6% versus 9.3% for the NHIS (Table 1) and 24.2% versus 10.3% for the NHANES (Table 2) for persons with versus without diabetes, respectively. In the BRFSS, prevalence of CHD or angina is 12.7% versus 5.2%, and for heart attack or MI, the prevalence is 13.5% versus 5.5% for those with and without diabetes, respectively (Table 3). The BRFSS is a cross-sectional, self-reported survey, which may limit interpretation. National data based on hospital discharges demonstrate similar trends between adults with and without diabetes (Table 4, Appendix A4).

Table Icon

TABLE 1.

Age-Standardized Prevalence of History of Heart Disease Among Adults Age ≥18 Years, by Diabetes Status, Sex, and Race/Ethnicity, NHIS, U.S., 2019–2020

Table Icon

TABLE 2.

Age-Standardized Prevalence of History of Heart Disease Among Adults Age ≥20 Years, by Diabetes Status, Sex, and Race/Ethnicity, NHANES, U.S., 2017–2020 Q1

Table Icon

TABLE 3.

Age-Standardized Prevalence of History of Heart Disease Among Adults Age ≥18 Years, by Diabetes Status, Sex, Race/Ethnicity, and Other Characteristics, BRFSS, U.S., 2019

Table Icon

TABLE 4.

Age-Standardized Prevalence of History of Heart Disease Among Adults Age ≥18 Years, by Diabetes Status, Sex, and Race, HCUP-NIS, U.S., 2018

Line graph showing that the prevalence of heart disease increases with age regardless of the presence of diabetes or sex.

FIGURE 2.

Prevalence of Coronary Heart Disease, by Diabetes Status, Age, and Sex, U.S., 2019–2020. Coronary heart disease and diabetes status are self-reported.

Examination of these national survey results demonstrates that the age-standardized prevalence of heart disease is consistently higher among men than women, regardless of diabetes status (Tables 1, 2, 3, and 4; Appendices A1, A2, A3, and A4). There is substantial variation in the age-standardized prevalence of heart disease based on race and ethnicity. Asian and American Indian/Alaska Native individuals with diabetes reported the highest rates of CHD or angina, and American Indian/Alaska Native persons with diabetes reported the highest rates of heart attack or MI in the BRFSS 2019 (Table 3). For the NHIS 2019–2020 and NHANES 2017–2020 Q1 (which did not have sufficient numbers of American Indian/Alaska Native respondents to report separately), non-Hispanic White individuals generally reported the highest rates of heart disease (Tables 1 and 2). However, non-Hispanic Black individuals in the NHANES reported more HF than individuals from other race/ethnicity groups (Table 2).

Importantly, sex differences exist for the relative risk of CHD in the presence of diabetes. In healthy young and middle-aged U.S. adults between ages 35 and 60 years, with different underlying risks of heart disease, a study across three diverse cohorts found that women with diabetes were significantly more likely to develop CHD than those without diabetes (pooled HR 3.61, 95% CI [confidence interval] 1.97–6.61), but similar relationships were not found in men (pooled HR 1.17, 95% CI 0.71–1.94), after adjustment for age, race, education, smoking, BMI, hypertension, and dyslipidemia (20). In a large meta-analysis of 22 prospective cohorts, an excess risk of fatal CHD was found in women (RR 3.12, 95% CI 2.34–4.17) and men (RR 1.99, 95% CI 1.69–2.35) with diabetes compared to those without diabetes; however, the ratio of relative risks was 1.46 (95% CI 1.14–1.88) comparing women to men. These results suggest that the relative risk of fatal CHD associated with diabetes is about 50% higher in women than men (11).

Interestingly, large secular trends have been noted from the 1970s to 2020. Figures 3 and 4 present the prevalence of diabetes and heart disease (self-reported heart attack or HF) for women and men, respectively, across six NHANES time periods covering 1976–2020. Over the same period, the prevalence of self-reported diabetes increased from 4% to 9% in women and from 3% to 12% in men. However, the prevalence of heart disease remained stable or decreased for men and women without diabetes but increased modestly in both men and women with diabetes. Although overall heart disease prevalence has been stable, population attributable risk (here defined as the number of cases of heart disease that would not occur if diabetes could be eliminated) may not have been. In other words, the prevalence of heart disease has remained stable, even though the prevalence of diabetes has increased—that is, the association between diabetes and heart disease has decreased, and the population attributable risk has declined.

Line graph showing trends in women between 1976 and 2020 which indicate that diabetes increased while heart disease remained stable.

FIGURE 3.

Trends in the Age-Standardized Prevalence of Diabetes and Heart Disease Among Adult Women Age ≥20 Years, U.S., 1976–2020 Q1. Diabetes is defined as self-reported or self-report and/or A1c (≥6.5%) and/or FPG (≥126 mg/dL); (more...)

Line graph showing trends in men between 1976 and 2020 which indicate that diabetes increased while heart disease remained stable.

FIGURE 4.

Trends in the Age-Standardized Prevalence of Diabetes and Heart Disease Among Adult Men Age ≥20 Years, U.S., 1976–2020 Q1. Diabetes is defined as self-reported or self-report and/or A1c (≥6.5%) and/or FPG (≥126 mg/dL); (more...)

One limitation of many national survey data is the reliance on self-reported diabetes and heart disease. However, as seen in Figures 3 and 4, the prevalence of diabetes based on self-report, FPG, and A1c levels has demonstrated the same trends as self-reported data alone over the last five survey time periods. Therefore, the reported variations by age, sex, and race/ethnicity are likely real.

Risk Factors for the Development of Heart Disease in People With Diabetes

Demographic Factors

Increasing age is a risk factor for development of heart disease in people both with and without diabetes (17). As noted, men with diabetes generally have a higher age-standardized prevalence of heart disease than women with diabetes in the United States, although the relative risk of heart disease in the presence of diabetes is higher in women compared to men.

Races/ethnicities at higher risk of heart disease in the presence of diabetes include American Indian/Alaska Native persons in the BRFSS (Table 3). Among participants in the NHANES I Epidemiologic Follow-up Study, the population attributable risk of CHD incidence associated with a medical history of diabetes was higher in Black women (8.7%) compared to White women (6.1%), suggesting that diabetes may confer differential risk of heart disease by race (21). Further, subgroups at higher risk of heart disease may exist within different races/ethnicities. As an example, grouping all Asians together may be misleading since South Asians have a higher risk of CVD than East Asians living in the United States and develop atherosclerotic cardiovascular disease (ASCVD) earlier in life; their ASCVD is more aggressive than in age-matched people of other ethnicities, with a fourfold higher risk of CVD than the general population (22).

Lifestyle Factors

Important major lifestyle factors that confer risk for CVD include physical activity and smoking, as well as diet and weight loss, which are also discussed in the Clinical Trials section.

Physical Activity

Most studies investigating the role of physical activity in the development of diabetes have been observational. The Health Professionals’ Follow-up Study (HPFS) (23) followed 2,803 men without physical impairment who reported a diagnosis of diabetes at age ≥30 years; men reported their physical activity every 2 years during 14 years of follow-up. Relative risks of CVD and death were estimated using Cox proportional hazards with adjustment for potential confounders. The multivariate relative risks of CVD incidence corresponding to quintiles of increasing total physical activity were 1.00 (reference), 0.87, 0.64, 0.72, and 0.67 (p for trend=0.07). The corresponding multivariate relative risks for total mortality were 1.00, 0.80, 0.57, 0.58, and 0.58 (p for trend=0.005). Walking was associated with reduced risk of total mortality, with relative risks across quintiles of walking of 1.00, 0.97, 0.87, 0.97, and 0.57 (p for trend=0.002). Further, walking pace was inversely associated with the risk of CVD, fatal CVD, and total mortality independent of walking hours.

The EPIC-Norfolk cohort study designed and tested a new four-part physical activity questionnaire, asking about (a) work-related physical activity; (b) leisure physical activity, including housework; (c) amount of energy expended during exercise based on sweating, rapid heart rate, and hours per week; and (d) stair climbing. Analyses based on these questions were derived from the responses of 4,423 men and 5,711 women, age 45–79 years (without CHD at baseline; 2% with diabetes) from 10 European countries, who had complete data on physical activity and CHD outcomes and were followed for an average of 10.9 years for fatal CHD (24). A total of 548 men and 310 women had a validated CHD event. Metabolic syndrome was diagnosed if at least three of five criteria were present (elevations in waist circumference, triglycerides, blood pressure, or A1c or low HDL-C). In both sexes, event rates were higher in those with the metabolic syndrome compared to those without: 17.4% versus 9.4% in men and 10.2% versus 3.4% in women. Significant downward trends in CHD event rates with increasing physical activity were strongest in men and women with the metabolic syndrome (37.6% of men and 30.2% of women). There was statistical evidence for significant effect modification (p for interaction=0.1 for men, p=0.06 for women, p=0.006 for both sexes combined), indicating that physical activity affected the association between CHD risk and the metabolic syndrome. Thus, interventions to increase physical activity targeting specific metabolic syndrome components are likely to decrease CHD risk in individuals with the metabolic syndrome.

Smoking

Overwhelming epidemiologic evidence has found that smoking cessation decreases the risk of CVD or CHD. The lifetime benefits of smoking cessation for persons with and without diabetes have been reported using the findings from meta-analyses of interventional and observational studies and then applying them to observational data from the Multiple Risk Factor Intervention Trial (MRFIT) cohort (25,26). In this cohort, it was estimated that smoking cessation would prolong the life of a 45-year-old man with diabetes by a mean of 3 years compared with 4 years for a 45-year-old man without diabetes and that the relative risk of CHD mortality for quitters compared to non-quitters was significantly lower (RR 0.63) after one year of smoking cessation. Using data from the Framingham Offspring Study (27), another study reported that recently quitting smoking was associated with an average weight gain of 2.7 kg in participants without diabetes and 3.6 kg in those with diabetes, but quitting still reduced CVD events by half. Results were stronger in the much larger group without a history of diabetes; among participants with diabetes, there were qualitatively similar lower risks that did not reach statistical significance, possibly because of limited study power.

Social Determinants of Health

The WHO defines the social determinants of health broadly as “the circumstances in which people are born, grow, live, work, and age, and the systems put in place to deal with illness” (28). In the context of diabetes and heart disease, these social determinants include socioeconomic status (i.e., income, education, employment, and other factors), race and ethnicity, social support, culture (including language), access to medical care, and residential environments (29). In data from the NHIS between 2011 and 2014 (30), increasing diabetes prevalence was found at lower levels of income. Using data from the NHANES 1999–2004 and the 2000 U.S. Census, individual poverty increased the odds of having diabetes for both White and Black persons (31). Further, low socioeconomic status is related to almost double the rate of CHD compared to those of high socioeconomic status, using a computer simulation model of CHD and stroke incidence, prevalence, and mortality among adults in the United States (32). Thus, social determinants of health are important contributors to the development of both diabetes and heart disease.

Metabolic Risk Factors

As a result of demographic, social, lifestyle, and/or genetic risk factors, “metabolic” risk factors act as the first measurable alert of increased CVD risk. Blood markers (of glycemia, lipoproteins, and inflammation), blood pressure, and BMI are all highly predictive of cardiovascular risk and are frequently comorbid. As a result, risk stratification schemes attempt to integrate as many metabolic factors into their risk estimates as possible (33,34).

Hyperglycemia and Insulin

Diabetes, as defined by measures and degree of hyperglycemia, is widely accepted as a high-risk state for CVD (35). Prior to the contemporary definitions for diabetes, the MRFIT cohort followed 347,978 men age 35–57 years at high risk for CHD for an average of 12 years to determine CVD mortality (36). Among 5,163 men who reported taking medication for diabetes (as the definition of diabetes), 1,092 men died; 603 of those deaths were due to CVD. Absolute risk for CVD death was three times higher for those with versus without diabetes even after adjustment for age, race/ethnicity, income, serum cholesterol, systolic blood pressure, and number of cigarettes smoked per day. The presumed conclusion from this study was that hyperglycemia was the major driver of CVD risk.

Following this presumption, much debate ensued as to whether diabetes defined by FPG compared to 2-hour post-challenge oral glucose tolerance test (OGTT) is more closely related to cardiovascular risk (14,37). The Diabetes Epidemiology: Collaborative Analysis of Diagnostic Criteria in Europe (DECODE) study group analyzed individual data from 22 European, 2 Japanese, and 1 American cohorts, which included 29,714 adults (without a diagnosis of diabetes) who were followed for an average of 11 years. The study was conducted to determine whether the glucose association with CVD was linear or showed a threshold effect and whether it was independent of classic CHD risk factors (38). In the pooled analysis for fatal CVD, a J-shaped association was found with a threshold effect for FPG (97.2 mg/dL [5.39 mmol/L]) and a linear association with 2-hour post-challenge glucose. Risk was increased at blood glucose levels less than those thought to be diagnostic of diabetes. This particular analysis found the 2-hour glucose was a stronger CVD risk factor than FPG.

With the declining popularity of the OGTT, the Emerging Risk Factors Collaboration conducted a meta-analysis of 102 prospective studies seeking to address uncertainties about the magnitude of associations of diabetes and fasting glycemia with the risk of CHD (17). Individual records from 698,782 people without known vascular disease who had 52,765 incident fatal or first-ever nonfatal vascular events during 8.49 million person-years at risk were included. Overall, the risk for CHD was about twofold greater in adults with diabetes at baseline compared to those without. These hazard ratios did not change appreciably after further adjustment for lipid, inflammatory, or renal markers. Hazard ratios for CHD were higher in women than in men, higher at age 40–59 years than at ≥70 years, and higher for fatal than nonfatal CVD. FPG had a U- or J-shaped association with vascular risk, with no significant associations in the normoglycemic range between 3.90 mmol/L (70.3 mg/dL) and 5.59 mmol/L (100.7 mg/dL). Compared with FPG concentrations of 3.90–5.59 mmol/L, hazard ratios for CHD were: 1.11 (95% CI 1.04–1.18) for 5.60–6.09 mmol/L (100.9–109.7 mg/dL) and 1.17 (95% CI 1.08–1.26) for 6.10–6.99 mmol/L (109.9–126 mg/dL), showing a significant, graded risk below FPG-defined diabetes (17).

In 2010, the American Diabetes Association (ADA) advocated the use of the A1c for diagnosis of both diabetes and prediabetes (39). Accordingly, interest in A1c as a marker of CVD risk ensued. Since this time, a number of population studies have demonstrated residual CVD risk across the A1c range after adjustment for known CVD risk factors (40,41,42), including below the threshold for A1c-defined diabetes. For example, after adjustment for age, sex, waist circumference, history of CVD, smoking, hypertension, and dyslipidemia in a large U.S. cohort, A1c 5.5%–6.0% (37–42 mmol/mol) versus A1c <5.5% was associated with a 25% higher risk of CHD and 16% higher risk of stroke that further increased to 88% and 119% higher for CHD and stroke, respectively, for A1c 6.0%–6.5% (42–48 mmol/mol) (42). Notably, the continuous relationship between A1c and CVD is not unique to the United States. Consistent findings have been reported in the United Kingdom (43), Australia (44), India (45), and in meta-analyses spanning a wide range of ethnic groups (46,47).

Given the limitations of the A1c to accurately measure glycemia in some populations, non-A1c markers of ambient glycemia (i.e., glycated albumin, 1,5-anhydroglucitol, and fructosamine) have gained some traction in clinical medicine. A post hoc analysis of the Atherosclerosis Risk in Communities (ARIC) study looked at the predictive value of these glycemic biomarkers for CVD in 10,373 participants followed from 1990–1992 until 2012 (48). Major findings revealed an association of all three—glycated albumin, 1,5-anhydroglucitol, and fructosamine—with CVD but only at their highest quintile blood concentrations and only in people with diabetes, whether diagnosed by FPG or A1c. No association was seen with these glycemic markers and CVD in people without diabetes, corroborating earlier findings (49).

The well-established fact that hyperglycemia, however measured, most often occurs in the setting of insulin resistance has led to speculation that insulin resistance, rather than hyperglycemia, drives CVD risk. Proving this hypothesis has been less than straightforward. Unlike glycemia, insulin resistance is difficult to measure outside of a research setting. Early reports linking hyperinsulinemia to CVD (50,51,52,53) were debunked by larger, contemporary trials controlling for confounding variables (54,55,56), although some disagreement remains (57). Many attribute contradictory findings to lack of standardization in the insulin assay, with many assays failing to discriminate between insulin and proinsulin or note whether the sample was taken in the fasted or fed state. Collectively, there is some indication that proinsulin, but neither fasting nor nonfasting insulin, associates with increased risk for CVD (58,59,60), albeit a marker of beta cell failure rather than insulin resistance. A single study using the intravenous glucose tolerance test (IVGTT) in conjunction with the minimal model demonstrated a 56% increase in coronary artery disease when comparing insulin sensitivity in the 75th versus 25th percentiles, even after adjusting for cardiovascular risk factors (56). The gold-standard test for assessment of insulin sensitivity—the hyperinsulinemic-euglycemic clamp—has yet to be tested as a predictor of CVD risk.

Lipids and Lipoproteins

Type 2 diabetes and insulin resistance are associated with abnormalities in circulating lipids, including elevated triglycerides and small, dense LDL particles, as well as low concentrations of HDL-C (61). These abnormalities in circulating concentrations of lipids often precede the diagnosis of type 2 diabetes (62,63,64,65). For a given concentration of LDL-C, the lipoprotein particles tend to be smaller and denser in patients with type 2 diabetes than in patients without diabetes (66).

Patients with type 2 diabetes tend to have higher triglyceride and lower HDL-C concentrations than patients without diabetes, and until recently, there has been disagreement as to whether triglycerides or HDL-C play a causal role in the development of ASCVD. Large observational studies have noted an association between triglyceride concentrations and cardiovascular events. In a comparison of individuals in the top third with those in the bottom third of usual log-triglyceride values, the adjusted odds ratio (OR) for CHD was 1.57 (95% CI 1.10–2.24) in the EPIC-Norfolk study and 1.76 (95% CI 1.39–2.21) in the Reykjavik study (67). However, the prevalence of diabetes at baseline in the two cohorts was low (2% and 3%, respectively). After adjusting for baseline values of established risk factors, including a history of diabetes, the strength of these associations was attenuated but remained statistically significant. Mendelian randomization studies are consistent with the hypothesis, but do not prove, that triglycerides or triglyceride-rich remnant lipoprotein particles cause atherosclerosis, even when accounting for LDL-C concentrations (68,69). As discussed in detail below, therapies to lower LDL-C concentrations have demonstrated important reductions in major adverse cardiovascular event (MACE) rates. Therapies directed at improving HDL-C and triglyceride concentrations have had more mixed success.

Hypertension

The CDC estimates that 69% of patients with type 2 diabetes have hypertension (19). Just as dyslipidemia can precede the diagnosis of diabetes, so too can the diagnosis of hypertension precede diabetes (62,70). As might be anticipated, patients with type 2 diabetes are also at elevated risk of developing hypertension (71). An epidemiologic analysis of data from the United Kingdom Prospective Diabetes Study (UKPDS) suggests an increase in both macrovascular and microvascular events with increases in systolic blood pressure in patients with diabetes, as well as a substantially elevated risk of HF and peripheral artery disease (PAD) (including amputation or death from PAD) (72).

These data have led the American Heart Association (AHA) to recommend a lower blood pressure treatment goal (<130/80 mmHg) in patients with diabetes with the goal of improving macrovascular and microvascular outcomes (73). However, recommendations about which patients to treat aggressively, and which to treat less aggressively, have historically differed among major professional organizations, although recent guidelines from the ADA reflect greater concordance with those from the AHA (74,75). Recommendations have differed in part because of concern about potential adverse effects of antihypertensive therapy in patients with diabetes and because of a lack of clarity from clinical trial data.

Obesity

Obesity was the first recognized and remains the strongest single risk factor for type 2 diabetes, as the obesity epidemic that began in the United States in the early 1980s was closely associated with the epidemic of diabetes (76). A subsequent epidemic of CVD was predicted, but thus far, the amount of CVD in the United States continues to decline overall in both sexes and in most race/ethnicity groups. The paradox of decreasing prevalence in CVD despite increasing prevalence of obesity—irrespective of diabetes status—has been attributed to improved lifestyle, use of better lipid-lowering and blood pressure medications, smoking cessation, and more revascularization surgery, as reviewed by Barrett-Connor (76).

Alternate hypotheses for the lack of increase in CVD prevalence despite the increase in obesity prevalence include uncertainties as to the measure of adiposity used in trials (i.e., BMI, waist circumference, or waist-to-hip ratio), lack of data on how change in any measure of adiposity predicts CVD events, and whether adiposity increases cardiovascular risk independent of frequently coexistent cardiovascular risk factors. BMI has been the most widely used measure of adiposity, when examining the association between adiposity and CVD (77), with separate “at-risk” cut points set for Caucasian and Asian populations (78), but it has also been the most openly criticized. One large, multinational case-control study found that waist-to-hip ratio was three times more strongly related to acute MI than was BMI (77). Other proponents advocate measuring waist circumference, citing its similar performance as waist-to-hip ratio in both the Nurses’ Health Study (79) and a combined analysis of 15 prospective cohort studies (80). Lack of agreement as to which measure is the correct measure of adiposity is further magnified when discussing the lack of data on how long-term change in each measure relates to CVD risk.

The Rancho Bernardo Cohort Study was among the first to prospectively examine the association between BMI and coronary artery calcium (CAC) burden (as a surrogate for subclinical CHD) (76). More recently, the Emerging Risk Factors Collaboration widened the scope, examining hazard ratios per 1-SD (standard deviation) higher baseline values (i.e., 4.56 kg/m2 higher BMI) across three measures of adiposity (81). In people with a BMI ≥20 kg/m2, hazard ratios for CVD were 1.23 (95% CI 1.17–1.29) per 1-SD higher BMI, 1.27 (95% CI 1.20–1.33) per 1-SD higher waist circumference, and 1.25 (95% CI 1.19–1.31) per 1-SD higher waist-to-hip ratio, after adjustment for age, sex, and smoking status. After further adjustment for baseline systolic blood pressure, diabetes history, and total and HDL cholesterol, corresponding hazard ratios were still significant at 1.07 (95% CI 1.03–1.11) with BMI, 1.10 (95% CI 1.05–1.14) with waist circumference, and 1.12 (95% CI 1.08–1.15) with waist-to-hip ratio. Reproducibility was greater for BMI than for waist circumference or waist-to-hip ratio, and findings were similar when adiposity measures were considered in combination. In a sub-analysis, addition of information on BMI, waist circumference, or waist-to-hip ratio to a CVD risk prediction model that included conventional risk factors did not improve risk discrimination. This sub-analysis led to the conclusion that BMI, waist circumference, and waist-to-hip ratio, whether assessed singly or in combination, do not independently improve CVD risk prediction in people in developed countries when additional information is available for systolic blood pressure, history of diabetes, and lipids.

In accordance with this conclusion, many consider obesity to be part of a “syndrome”—Syndrome X, Insulin Resistance Syndrome, or the Metabolic Syndrome—rather than an independent risk factor for CVD. Popularized in the 1980s (82), much ado has been made about the definition that predicts cardiovascular risk. The initial choice of the components—hypertension, high triglycerides, low HDL-C levels, waist girth, or waist-to-hip ratio, and in some definitions, high insulin levels or diabetes—was logical, but the cut points defining normality were arbitrary. Interestingly, definitions put forth by the National Cholesterol Education Program (Adult Treatment Panel III) (NCEP) (83), the International Diabetes Federation (IDF) (84) and the WHO (85) have all been shown to predict incident type 2 diabetes and CVD, although it is unclear which one is best (86,87). Controversy over the scientific basis for the definition of risk factors and their clustering stems from the arbitrariness of the included (and excluded) variables and their cut points, the loss of risk prediction when continuous risk factors are categorized, and the unproven assumption that there is a single underlying biology (88,89). Ultimately, unresolved controversy combined with the observation that the metabolic syndrome was inferior to established risk factor models like the Framingham CVD risk score for either type 2 diabetes or CVD in the NHANES cohort (90) has diminished its popularity as a distinct entity.

Despite the fact that the “metabolic syndrome” may have lost its limelight, the concept remains important. A number of legacy trials for diabetes prevention have shown that interventions targeting weight reduction—with collateral benefits on blood triglycerides, HDL-C, glycemia, and blood pressure—have beneficial effects on microvascular, macrovascular, and/or mortality endpoints. The U.S. Diabetes Prevention Program Outcomes Study (DPPOS) showed a 28% lower prevalence in the composite microvascular endpoint after 15 years for those who did not develop diabetes (both active interventions had favorable effects on components of the metabolic syndrome vs. placebo) (91). Most notable was the 30-year data from the Da Qing Study showing a 26% reduction in cardiovascular events, 33% reduction in cardiovascular mortality, and 26% reduction in all-cause mortality (92) in participants previously receiving a lifestyle intervention. Participants in these trials had impaired glucose tolerance (or prediabetes) at baseline, most (~85%) of whom also had metabolic syndrome. People with established type 2 diabetes who lost >10% of their body weight in the first year of the Look AHEAD: Action for Health in Diabetes study had a 21% and 24% reduction in the composite primary and secondary cardiovascular outcomes, respectively (93). Nevertheless, whether the benefits on cardiovascular outcomes specifically relate to weight loss or the combined reduction in cardiovascular risk factors is unclear. The latter is widely hypothesized to be the reason why the DPPOS did not show a lower prevalence in CVD after 21 years of follow-up (94).

Novel Risk Markers

A biomarker is a biological marker that indicates the presence of a disease or the risk for a disease state. Clinically useful biomarkers are ones that can be measured, improve diagnostic or prognostic performance, and aid in the clinical management of patients by guiding the initiation, duration, or intensity of therapy. As imaging biomarkers (i.e., subclinical atherosclerosis) are discussed in another section, this section focuses on circulating blood biomarkers, excluding lipid biomarkers and glycemic markers, which are also discussed in other sections. A large number of biomarkers have been examined for their utility in CVD risk assessment, but for space limitation, only the ones most commonly used for CVD risk assessment are described, including hsCRP, high-sensitivity cardiac troponin (hs-cTn), B-type natriuretic peptides (BNP), and adipokines.

C-Reactive Protein

C-reactive protein is a marker of inflammation, and hsCRP levels are higher among individuals with obesity, insulin resistance, and diabetes. Among women without diabetes, higher CRP levels are independently associated with fasting insulin levels (95). Furthermore, hsCRP levels predict the risk for incident type 2 diabetes (96,97,98), implicating the role of low-grade inflammation in diabetes pathogenesis. However, associations of inflammation with diabetes risk have been attenuated after accounting for BMI (97,98), suggesting adiposity may be mediating this relationship.

In addition to predicting future type 2 diabetes risk, elevated levels of hsCRP predict incident ASCVD and improve risk prediction independently of the lipid profile (99,100). The Reynolds Risk Scores for prediction of CVD, which incorporate hsCRP in their equations, improve risk discrimination over traditional risk factors alone (101,102).

The Justification for the Use of Statins in Prevention: An Intervention Trial Evaluating Rosuvastatin (JUPITER) enrolled apparently healthy individuals without CVD or diabetes, who had LDL-C levels <130 mg/dL (<3.37 mmol/L) but elevated hsCRP levels ≥2 mg/L, and found that statin treatment reduced MACE by 44% (HR 0.56, 95% CI 0.46–0.69) compared to placebo (103). This pivotal trial provided key evidence demonstrating the benefit of statin therapy in patients with elevated hsCRP levels. Although this study excluded persons with diabetes, 41% of participants did have the metabolic syndrome, and the benefit of statin therapy was seen across metabolic syndrome groups. Therefore, the 2019 American College of Cardiology (ACC)/AHA primary prevention guideline considers elevated hsCRP ≥2 mg/L to be a “risk-enhancing” factor that would favor the initiation of statin therapy among individuals at borderline or intermediate risk. However, as persons with diabetes who are age ≥40 years are already recommended for statin therapy, the role of hsCRP assessment for guiding treatment decisions in this population is unclear.

High-Sensitivity Cardiac Troponin

Cardiac troponin is a protein found in cardiac myocytes that is released in the setting of myocardial injury (104). Although its clinical utility is established in the diagnosis of MI, its utility in predicting risk of incident ASCVD risk among asymptomatic individuals has been increasingly recognized (105). The development of high-sensitivity troponin assays (i.e., hs-cTnT and hs-cTnI) permits the detection of circulating troponin at much lower thresholds, on the order of ng/L, as early markers of subclinical myocardial injury (106).

In patients with type 2 diabetes and chronic, stable coronary artery disease who were enrolled in the Bypass Angioplasty Revascularization Investigation 2 Diabetes (BARI-2D) trial, hs-cTn concentration was an independent predictor of death from cardiovascular causes, MI, or stroke; however, among patients with abnormal hs-cTn concentrations, random assignment to prompt revascularization compared with medical therapy alone did not result in a significant reduction in the rate of the composite cardiovascular endpoint (107). In general population cohorts, elevated levels of hs-cTn have been independently associated with future ASCVD events even after accounting for traditional CVD risk factors (108,109). The addition of hs-cTn improves risk prediction compared to other risk prediction equations, such as the pooled cohort equations (PCE) and the Framingham Risk Score (109,110). Furthermore, in a primary prevention clinical trial, statin therapy significantly reduced hs-cTn over 5 years compared to placebo, and participants who experienced the greatest reduction in hs-cTn had a fivefold lower risk of coronary events compared to those whose hs-cTn increased (110).

Cardiac troponin also predicts risk among individuals with established diabetes. Among middle-aged adults with diabetes in the ARIC study, subclinical elevation of hs-cTn was independently associated with increased risk of incident ASCVD, HF, and all-cause mortality (111). In fact, persons with diabetes with hs-cTn above the 90th percentile had similar risks as individuals with established CVD (112). This finding suggests that the use of hs-cTn can help improve risk stratification in persons with diabetes and potentially guide clinical management, such as intensification of preventive therapies. Nevertheless, current guidelines do not specifically endorse or recommend the use of hs-cTn in risk assessment or management of asymptomatic individuals.

B-Type Natriuretic Peptides

BNPs are secreted from cardiac myocytes in response to myocardial wall stress, which can occur in the setting of volume expansion and/or pressure overload (113). While normally BNP plays a favorable physiologic role by promoting vasodilation and natriuresis, its elevation in the blood signals compensatory adaptation to a pathological state, such as subclinical or clinical HF, left ventricular hypertrophy, or myocardial ischemia (114,115,116,117). ProBNP is the prehormone that is cleaved to the active hormone BNP and also to the N-terminal pro B-type natriuretic peptide (NT-proBNP). Both BNP and NT-proBNP levels are used clinically in the evaluation of patients with suspected HF. However, even among asymptomatic individuals free of clinical ASCVD or HF, elevated NT-proBNP levels are associated with an increased risk of incident CVD (118), hospitalizations for HF (119), and cardiovascular mortality (120,121). In patients with established diabetes, higher levels of BNP predict future CVD and mortality (111,122,123). Interestingly, higher NT-proBNP and BNP levels are associated with lower risk of incident diabetes, which actually suggests a favorable role of natriuretic peptides for type 2 diabetes prevention (124,125,126). Mendelian randomization studies are consistent with the hypothesis that the BNP locus may have a causal role in the development of diabetes (127). Similar to hs-cTn, current guidelines do not specifically endorse or recommend the use of natriuretic peptide measurement for risk assessment in asymptomatic individuals (75). However, this biomarker does have utility in the diagnosis of HF (128). Of note, BNP levels are paradoxically lower in persons with obesity compared to those without obesity, adding to the challenge of diagnosing HF with preserved ejection fraction among individuals with obesity (129,130).

Adipokines

Adipokines, such as leptin, adiponectin, and resistin, are endogenous hormones that are released from adipose tissue and influence several metabolic processes, including insulin sensitivity, endothelial function, and appetite regulation (131). Proinflammatory adipokines promote inflammation and insulin resistance, whereas anti-inflammatory adipokines appear to have a protective role. Adverse levels of adipokines may predict risk of incident type 2 diabetes (132). Studies have identified an association between an adverse adipokine profile with incident diabetes (133) and ASCVD (134,135,136,137,138).

Adiponectin, a favorable adipokine, was associated with decreased risk for type 2 diabetes in a large meta-analysis (132), but its association with ASCVD was not as robust (139). Leptin, an unfavorable adipokine, has been associated with increased risk of incident diabetes (140,141); however, the association of leptin with incident CVD has been mixed, with some studies showing increased risk, while other studies found the association to be attenuated after adjusting for BMI (142,143). Currently, adipokine levels are not routinely measured as part of clinical management or risk assessment.

Subclinical Atherosclerosis

There is significant heterogeneity in the risk for ASCVD events among individuals with diabetes, and it has become clear that the presence of diabetes by itself is not a CHD risk equivalent (144). Thus, among individuals with diabetes but without clinical ASCVD, there sometimes can be uncertainty regarding risk, and subclinical atherosclerosis imaging can be a useful tool to more accurately risk stratify individuals and guide shared decision-making around the use of preventive pharmacotherapies, such as statins and aspirin (145,146).

The CAC score, as measured by noncontrast cardiac computed tomography (CT), is a surrogate measure of total coronary atherosclerosis burden. CAC presence and the burden of CAC prognosticate future risk of ASCVD events in graded fashion, independently of age and other cardiovascular risk factors (147). CAC has emerged as the superior marker of risk and more accurately reclassifies risk compared to other subclinical markers, such as carotid intimal medial thickness (cIMT), ankle-brachial index (ABI), brachial reactivity, and hsCRP (148,149). In addition to CAC’s role in upgrading risk when present (i.e., CAC >0) and identifying individuals who might benefit from more intensive preventive therapy, perhaps even more importantly, the absence of CAC (i.e., CAC score=0) can downgrade an individual’s risk into a lower risk category where potentially statin therapy could be deferred or postponed if that is the patient’s preference (150).

In the Multi-Ethnic Study of Atherosclerosis (MESA), the CAC score strongly discriminated ASCVD risk among individuals with diabetes. Persons with type 2 diabetes with CAC scores ≥100 generally had ASCVD event rates of >20 per 1,000 person-years, whereas persons with type 2 diabetes who had A1c <7% (<53 mmol/mol) and CAC=0 had much lower ASCVD event rates of 4.5 per 1,000 person-years (151). Furthermore, the CAC score remained strongly prognostic of risk for incident CHD and ASCVD in multivariable models adjusted for traditional risk factors, diabetes duration of ≥10 years, use of insulin, and glycemic control (151). In the CAC Consortium study, the CAC score predicted CHD, CVD, and all-cause mortality among individuals with diabetes, and while women with diabetes had a lower prevalence of CAC compared to men with diabetes, a greater CAC score predicted CVD and mortality more strongly in women compared to men (152).

The presence of CAC, and particularly high scores >100, also predict risk of subsequent CVD among individuals with type 1 diabetes, with CAC scores >100–300 and >300 being associated with a fourfold and fivefold increased risk of CVD, respectively, even after adjustment for traditional risk factors and A1c (147). Whereas in that same cohort, CVD event rates among individuals with type 1 diabetes and CAC=0 were low (5.6 per 1,000 patient years) (147).

Note that while a CAC score of zero is associated with lower ASCVD risk, that certainly does not mean “no risk,” and clinical judgement should prevail. Among individuals with CAC=0, the presence of diabetes was associated with increased risk of ASCVD over 16 years of follow-up (153). Thus, the absence of CAC among patients with diabetes may identify low short-term risk, but not low long-term risk. In another epidemiology cohort study, among asymptomatic individuals with diabetes, a CAC score of zero was associated with low 5-year risk of mortality, similar to individuals without diabetes, but the risk among those with diabetes was greater after 5 years (154). Therefore, individuals with diabetes with a CAC score initially of zero may benefit from re-imaging, if preventive pharmacotherapy is not already initiated, to evaluate for any interim progression of disease and to reclassify risk. A prior analysis from MESA suggested that the warranty period of a CAC=0 (defined as the interval when the CAC scan should be repeated) may be shorter for individuals with diabetes compared to those without diabetes (3.4 vs. 6.4 years) (155), and thus, approximately 3 years may be time for rescanning if decisions remain uncertain about a patient’s preventive management.

Individuals with diabetes have a greater rate of CAC progression compared to individuals without diabetes, and CAC progression also predicts ASCVD events (156). Progression of CAC is more likely when diabetes control is suboptimal (157). Yet, patients with diabetes with more favorable cardiovascular risk profiles can remain free of progression. In a MESA study of individuals who were free of ASCVD at baseline, more than 40% of participants with diabetes remained free of CAC (i.e., CAC=0) over the 10-year period; among these participants, a more optimal cardiovascular risk profile and the absence of baseline atherosclerosis in other territories (i.e., carotid plaque = 0, thoracic aortic calcium = 0) were associated with greater odds of remaining free of incident CAC (158).

Treatment: Control of Glycemia or Other CVD Risk Factors and CVD Outcomes

Trials of Intensive Glycemic Control

Extensive epidemiologic evidence showing an association between diabetes and CVD (17) led to the hypothesis that treating diabetes to reduce blood glucose levels would lower CVD risk as a primary or secondary outcome (Table 5). The first study to test this hypothesis was the DCCT, which enrolled 1,441 people age 13–39 years with recent-onset type 1 diabetes and randomized them to either intensive (≥3 insulin injections per day—or via insulin pump—dose-adjusted to achieve a fasting glucose 70–120 mg/dL [3.89–6.66 mmol/L] and postprandial glucose <180 mg/dL [<10.00 mmol/L]) or conventional (one or two insulin injections per day dose-adjusted to avoid ketonuria or symptoms of hyperglycemia or glycosuria) insulin therapy for a mean follow-up of 6.5 years. Findings from this trial were the first to conclusively show clear benefit from intensive versus conventional glycemic control in reducing the development of retinopathy by 76%, microalbuminuria by 39%, and clinical neuropathy by 60% (159). While 41% fewer CVD events occurred in the intensively treated group, this number was not statistically significant, possibly because of the young age of the participants or small number of events.

Table Icon

TABLE 5.

Clinical Trials of Intensive Glucose Control on Cardiovascular Disease Risk

Examination of whether intensive glycemia control could prevent complications, including CVD, in people with type 2 diabetes then became a topic of inquiry in the UKPDS, which enrolled 3,867 adults with newly diagnosed type 2 diabetes and randomized them to either sulfonylurea or insulin versus diet alone for a mean follow-up of 10 years (160). Mean A1c levels of 7.0% and 7.9% (63 mmol/mol) were achieved in the drug versus diet-treated groups, respectively, and microvascular events were reduced 21%–34% by drug treatment. Although a metformin-treated subgroup (n=342) had 42% less diabetes-related death and 36% less all-cause mortality compared to the diet-treated subgroup (n=411) (161), the main trial failed to show a significant impact of intensive control on their composite CVD outcome (16% reduction, p=0.052) (161).

The near miss of the UKPDS to show CVD benefit led to widespread speculation that larger studies of people with type 2 diabetes at higher cardiovascular risk may benefit with greater intensification of their A1c than had been attempted in the UKPDS. Three large trials ensued—Action in Diabetes and Vascular Disease: Preterax and Diamicron Evaluation (ADVANCE) (162), Action to Control Cardiovascular Risk in Diabetes (ACCORD) (163), and the Veterans Affairs Diabetes Study (VADT) (164)—and were published in rapid succession. ADVANCE enrolled 11,140 adults age ≥55 years with type 2 diabetes with an additional cardiovascular risk factor and randomized them to glucose-lowering medications to achieve an A1c <6.5% versus standard care for a median follow-up of 5 years (162). Again, microvascular events were reduced by 14% (in this case, driven by less microalbuminuria) with no reduction in macrovascular events.

More concerning than the lack of cardiovascular benefit observed was the increase in all-cause mortality reported in ACCORD. ACCORD enrolled 10,251 people with type 2 diabetes with an average age of 62 years and randomized them to intensive glucose-lowering medications to achieve an A1c <6.0% versus A1c 7.0%–7.9% for a mean follow-up of 3.5 years (163). The trial was stopped early due to an observed 22% increased rate of all-cause mortality. Lastly, the VADT enrolled 1,791 military veterans with type 2 diabetes, 40% of whom had already had a cardiovascular event, and randomized them to glucose-lowering medications to achieve an A1c reduction of 1.5% greater than the control group for a mean follow-up of 5.6 years (164). Although benefit for several renal parameters was reported, no benefit was seen for the composite cardiovascular endpoint or any of the individual cardiovascular measures.

The concept that glucose lowering reduced cardiovascular events in people with diabetes was virtually abandoned. Fortunately, several of these trials entered long-term observational phases that have provided valuable lessons. The DCCT morphed into the Epidemiology of Diabetes Interventions and Complications (EDIC). During the 30-year follow-up of participants in EDIC, intensive treatment (~25 years post-intervention end) rendered a 32% reduction in cardiovascular events, lending credence to the notion of “metabolic memory” (165). Ten-year follow-up of the UKPDS boasted a persistent 24% reduction in microvascular disease and, finally, a significant 15% lower rate of MI and 15% less all-cause mortality (166). Unfortunately, after 15 years, the VADT did not demonstrate benefits in observational follow-up, as the A1c values had long since converged between groups (167).

A large meta-analysis combined 13 trials all aimed at showing the benefit of tight glycemic control (i.e., A1c <7%) for CVD prevention in people with type 2 diabetes, including the landmark trials described above (168). When combined and including 34,533 participants with varying durations of diabetes, intensive glycemic control was associated with a 15% reduction in nonfatal MI, with no benefit for all-cause or cardiovascular mortality compared to the standard glucose-lowering group (as defined in each trial), confirming its modest importance for cardiovascular protection over the short term in an out-patient setting.

Concurrently, a number of studies examined the benefit of glycemic control in the inpatient setting. Trials examining the benefit of insulin therapy post-MI yielded equivocal results (Diabetes Intensive Glucose After Myocardial Infarction [DIGAMI] 1 & 2 (169,170)), and the benefit of insulin therapy in a surgical intensive care unit (171) could not be reproduced in a medical intensive care unit (172). To reconcile the disagreements, the larger Normoglycemia in Intensive Care Evaluation – Survival Using Glucose Algorithm Regulation (NICE SUGAR) study randomized 6,104 critically ill patients requiring 3 or more days of hospitalization in an intensive care unit to intensive versus conventional glucose control (blood glucose 81–108 mg/dL [4.50–5.99 mmol/L] vs. <180 mg/dL) (173). Major findings from this trial showed no benefit (in median number of days in the hospital, intensive care unit, or requiring mechanical ventilation, or the need for renal replacement therapy), yet clear detriment (more severe hypoglycemia and higher 90-day mortality), in the intensively treated patients.

Lipid-Modifying Clinical Trials

LDL-C is the main etiologic factor for the development and progression of ASCVD, with overwhelming evidence from genetic, population, and interventional data supporting the causal role of LDL-C in ASCVD pathogenesis (174,175). Therefore, LDL-C reduction is a central tenet of ASCVD treatment and prevention for both primary and secondary patients, with the recommended intensity of therapy generally matched to the absolute risk of the patient (175). Moreover, ASCVD risk reduction is proportional to the magnitude of LDL-C lowering.

Statins

Statins block the synthesis of cholesterol in the liver by inhibiting the enzyme HMG Co-A reductase, which leads to up-regulation of the LDL receptor and a decrease in circulating LDL-C levels. In a meta-analysis of statin trials, among 18,686 individuals with diabetes (compared to 71,370 without diabetes), statin therapy conferred a similar 21% reduction in MACE for persons with and without diabetes (RR 0.79, 95% CI 0.72–0.86 and RR 0.79, 95% CI 0.76–0.82, respectively) (176). For every 1,000 persons with diabetes treated with statin therapy over 5 years, 42 (95% CI 30–55) fewer individuals had major vascular events compared to placebo-treated patients. Notably, the benefit of statin therapy was similar among persons with diabetes for both primary and secondary prevention (176).

Statins, particularly high-intensity statins, do have a small risk of new-onset diabetes (177) and potentially worsen glycemic control in an LDL-C reduction-dependent manner (178). New-onset diabetes conferred by statin therapy predominantly is seen in patients who already have risk factors for or evidence of glucose intolerance, such as elevated BMI or prediabetes (179). However, as mentioned, statins significantly reduce vascular events among individuals with diabetes, so the ASCVD benefits outweigh the small increase in blood glucose. Lifestyle changes and weight management, along with statin initiation, are recommended to offset the small rise in blood glucose.

Ezetimibe

Ezetimibe is an oral agent that blocks cholesterol absorption in the small intestine. 2018 AHA/ACC guidelines recommend the addition of ezetimibe to maximally tolerated statins in high-risk patients if the LDL-C remains above a threshold of >70 mg/dL (>1.81 mmol/L) (179). The IMPROVE IT trial (Improved Reduction of Outcomes: Vytorin Efficacy International Trial) found among >18,000 high-risk patients with acute coronary syndrome (ACS) that the addition of ezetimibe to statin (compared to statin alone) further reduced MACE over 7 years of follow-up. In the IMPROVE IT trial, 4,933 patients had diabetes and experienced greater absolute and relative risk reductions with ezetimibe (absolute risk reduction 5.5%; HR 0.85, 95% CI 0.78–0.94) compared to patients without diabetes (absolute risk reduction 0.7%; HR 0.98, 95% CI 0.91–10.4), p-interaction for diabetes status=0.02 (180). This finding suggests that the benefit of adding ezetimibe to a statin was even greater among high-risk patients with diabetes compared to those without.

PCSK9 Inhibitors

Proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i) block the PCSK9 protein in the liver involved in the degradation of the LDL receptor; this inhibition leads to upregulation of the LDL receptor on the surface of the liver and increased clearance of LDL-C from circulation.

The monoclonal antibody PCSK9i, evolocumab or alirocumab, are generally dosed every 14 days or monthly by subcutaneous injection and can reduce LDL-C by approximately 60%. Two large CVOTs demonstrated the benefit of a monoclonal antibody PCSK9i on top of maximally tolerated statin therapy in high-risk patients with ASCVD—the FOURIER trial (Further Cardiovascular Outcomes Research With PCSK9 Inhibition in Subjects With Elevated Risk) with evolocumab (181) and the ODYSSEY Outcome trial (Evaluation of Cardiovascular Outcomes After an Acute Coronary Syndrome During Treatment With Alirocumab) with alirocumab (182). A sub-analysis of FOURIER showed that evolocumab therapy significantly reduced MACE in patients with and without diabetes in a similar fashion, without effect modification by diabetes status (HR 0.83, 95% CI 0.75–0.93 for patients with diabetes and HR 0.87, 95% CI 0.79–0.96 for patients without diabetes; p-interaction=0.60) (181). Notably, evolocumab in FOURIER did not increase the risk of new-onset diabetes or worsen hyperglycemia. The ODYSSEY Outcome trial, which included even higher-risk patients with recent ACS, demonstrated similar relative risk reductions in MACE for each glycemic category but greater absolute risk reduction with alirocumab for patients with diabetes (2.3% reduction, 95% CI 0.4%–4.2% vs. 1.2%, 95% CI 0.0%–2.4% in prediabetes and 1.2%, 95% CI -0.3%–2.7% in normoglycemia, p-interaction=0.0019) (182). Similar to the FOURIER trial, PCSK9 inhibition in the ODYSSEY Outcome trial was not associated with increased risk of new-onset diabetes or worsening glycemia. These data confirm the safety and efficacy of PCSK9i therapy in persons with diabetes.

Inclisiran is a small interfering RNA (siRNA) molecule that prevents the translation of the PCSK9 protein. After baseline and 3-month doses, inclisiran is then dosed every 6 months by subcutaneous injection, which may represent an important strategy for improving adherence and sustained LDL-C reduction in high-risk patients. In pooled patient-level data from three phase 3 trials (ORION 9, ORION 10, and ORION 11) that included patients with ASCVD and/or familial hypercholesterolemia (36% with diabetes), inclisiran lowered LDL-C by approximately 50% (183). Similar treatment effect of inclisiran was seen regardless of baseline diabetes status (184,185). Inclisiran is approved by the U.S. Food and Drug Administration (FDA) for patients with ASCVD who need additional LDL-C lowering after maximal tolerated statin. The CVOT evaluating inclisiran (ORION-4, NCT03705234) is currently ongoing.

Bempedoic Acid

Bempedoic acid is a newer oral agent for LDL-C lowering that works by blocking an enzyme (ATP citrate lyase) involved in cholesterol synthesis in the pathway upstream from HMG-CoA reductase (the target of statins). The CLEAR (Cholesterol Lowering via Bempedoic Acid, an ACL-Inhibiting Regimen) clinical trials have demonstrated similar efficacy of LDL-C lowering in patients with and without diabetes (about -18% lower LDL-C compared to placebo) (186). Notably, unlike statins, bempedoic acid is associated with a lower risk of developing new-onset diabetes or worsening glycemia in a meta-analysis (OR 0.66, 95% CI 0.48–0.90) (186,187). The CVOT of bempedoic acid, in which 46% of participants had diabetes, showed that bempedoic acid reduced LDL-C by 22% and 4-point MACE by 13%, with similar relative risk reduction among persons with and without diabetes (188).

Icosapent Ethyl

Icosapent ethyl (IPE) is a highly purified form of eicosapentaenoic acid (EPA), an omega-3 fatty acid. In the REDUCE-IT (the Reduction of Cardiovascular Events with Icosapent Ethyl–Intervention Trial), IPE significantly reduced MACE by an impressive 25% among patients with established ASCVD or with diabetes who had at least one other risk factor and moderate hypertriglyceridemia (135–500 mg/dL [1.53–5.65 mmol/L]) despite statin treatment and controlled LDL-C (189). The benefits on MACE reduction with IPE exceeded what would be anticipated by the 17% reduction in triglycerides alone, and likely, the benefit of IPE was conferred through additional mechanisms, such as its anti-thrombotic and anti-inflammatory properties, independent of triglyceride-lowering effects. In REDUCE-IT DIABETES subgroup analysis, among the 4,787 patients with diabetes, IPE conferred a 7% absolute and 23% relative risk reduction (RR 0.77, 95% CI 0.66–0.88) in MACE, without any significant change in A1c or glucose (190). It should be noted that other omega-3 preparations, typically docosahexaenoic acid (DHA) combined with EPA, even at high doses, have not been demonstrated to similarly reduce cardiovascular events (191,192,193). Differences in the cardiovascular benefits of omega-3s are not fully understood but are likely due to differences in biological properties of DHA and EPA in membrane stabilization, antioxidant, and other cardioprotective effects, as well as the achieved serum EPA levels necessarily to confer benefit. A small increased risk of atrial fibrillation is seen with high-dose omega-3 preparations (189,192,194). Fortunately, this did not translate into increased risk of ischemic stroke in REDUCE-IT (stroke risk was lower in the IPE arm) (189).

Fibrates

Fibrates were one of the earliest classes of medications used to reduce triglyceride levels. Fibrates work by modulating peroxisome proliferator-activated receptor alpha (PPAR-α) and, as such, influence the expression of apolipoprotein C-III, apolipoprotein A-I, lipoprotein lipase, and other proteins involved in lipid metabolism.

The 1999 Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial (VA-HIT) was a secondary prevention trial of men with CHD and low HDL-C (25% with diabetes) and demonstrated that gemfibrozil reduced the risk of MACE, but these patients were not treated with background statin therapy (195). The Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) and the ACCORD Lipid study also evaluated the role of fibrates in ASCVD risk reduction. The FIELD trial randomized nearly 10,000 participants with diabetes, but not on statin therapy, to receive fenofibrate versus placebo (196). Fenofibrate reduced nonfatal MI and revascularization events but failed to demonstrate a statistically significant difference in the primary composite outcome of first MI or CHD death. The ACCORD Lipid substudy then evaluated fenofibrate for residual risk reduction in patients already on a background of statin therapy (197). However, there was no difference in the primary outcome of nonfatal MI, nonfatal stroke, or cardiovascular mortality in patients receiving fenofibrate plus simvastatin compared to those receiving simvastatin alone. The Pemafibrate to Reduce Cardiovascular Outcomes by Reducing Triglycerides in Patients With Diabetes (PROMINENT) trial assessed the role of pemafibrate as add-on therapy in reducing residual ASCVD risk in statin-treated patients with mild-to-moderate hypertriglyceridemia (198). The study was stopped due to futility; the incidence of cardiovascular events was not lower among those who received pemafibrate than among those who received placebo. Thus, to date, no trials have demonstrated a cardiovascular outcome benefit of fibrates when added to statins. However, fibrates still have a role in the treatment of severe hypertriglyceridemia to reduce the risk of pancreatitis.

Trials of Blood Pressure Control

Reducing blood pressure, usually with pharmacologic therapy, is one of the cornerstones of CVD prevention. A few trials of patients with type 2 diabetes specifically sought to address the question of whether a blood pressure target of <130/80 mmHg, or lower, was better than the standard target of <140/90 mmHg. The trials and their results are outlined in Table 6. The Hypertension Optimal Treatment (HOT) trial, published nearly 25 years ago, included 1,501 patients with type 2 diabetes out of a total sample of 18,790 and randomized patients to a goal diastolic blood pressure of ≤80 mmHg, ≤85 mmHg, or ≤90 mmHg (199). For all patients randomized to ≤80 mmHg compared to those randomized to ≤90 mmHg, regardless of diabetes status, no significant reduction was seen in the risk of nonfatal MI, nonfatal stroke, or cardiovascular death (3-point MACE; RR 0.93, 95% CI 0.78–1.12). However, in the subset of 1,501 patients with type 2 diabetes, a significant reduction in 3-point MACE was seen (RR 0.49, 95% CI 0.29–0.81). The ADVANCE BP trial enrolled 11,140 patients with type 2 diabetes and randomly allocated them in a 1:1 ratio to fixed-dose perindopril/indapamide or placebo (200). The achieved mean blood pressure was 136/73 mmHg in the active treatment group and 142/75 mmHg in the placebo group. Significant reduction was observed in the trial primary endpoint of cardiovascular death, MI, stroke, or microvascular (new or worsening renal or diabetic eye disease) events (HR 0.91, 95% CI 0.83–1.00, p=0.04). The 3-point MACE outcome showed a similar direction and magnitude of effect but was not statistically significant (HR 0.92, 95% CI 0.81–1.04). The ACCORD BP trial randomized 4,733 patients with type 2 diabetes in a 1:1 ratio to a target systolic blood pressure of <120 mmHg (intensively managed group) compared to <140 mmHg (201). There was no significant difference in the primary composite endpoint of time to a first nonfatal MI, nonfatal stroke, or cardiovascular death, with an event rate of 1.9% in the intensive target arm and 2.1% in the standard target arm (HR 0.88, 95% CI 0.73–1.06) (202).

Table Icon

TABLE 6.

Randomized Controlled Trials of Intensive Blood Pressure Control That Included Patients With Type 2 Diabetes

However, evidence from the Systolic Blood Pressure Intervention Trial (SPRINT) trial has reenergized this debate, even though SPRINT specifically excluded patients with diabetes. To what extent SPRINT trial findings can be extended to patients with type 2 diabetes is not clear. Eligible patients randomized to a goal systolic blood pressure of <120 mmHg compared to <140 mmHg had a 25% relative reduction in the risk of the primary composite outcome of nonfatal MI, ACS, stroke, HF, or cardiovascular death (HR 0.75, 95% CI 0.64–0.89) (203). Data from a trial conducted in China, in which patients age 60–80 years were randomly allocated to an intensive blood pressure control arm (goal systolic blood pressure 110–<130 mmHg) or a standard control arm (goal systolic blood pressure 130–<150 mmHg), found a substantial reduction in the primary composite outcome of stroke, ACS (acute MI and hospitalization for unstable angina), acute decompensated HF, coronary revascularization, atrial fibrillation, or death from cardiovascular causes (HR 0.74, 95% CI 0.60–0.92) (204). Approximately 19% of the 8,511 patients randomized had type 2 diabetes at baseline, and no evidence of heterogeneity of the benefits of intensive therapy was found when the trial was stratified by the presence or absence of diabetes at baseline (204).

Trials of Antiplatelet/Aspirin Management

Patients with diabetes are at increased risk for atherothrombosis due to hyperreactive platelets, hypercoagulable state, and endothelial dysfunction (205). Thus, aspirin therapy has been extensively explored as a way to mitigate ASCVD risk among persons with diabetes.

Aspirin is a well-established therapy for the secondary prevention of ASCVD among individuals with a prior history of CHD, transient ischemic attack (TIA)/stroke, and/or PAD. Aspirin reduces the risk of atherothrombosis by irreversibly inhibiting platelet function, but it also increases risk of bleeding by this same mechanism. Aspirin remains indicated for secondary prevention, where it confers a 19% reduction in serious vascular events (RR 0.81, 95% CI 0.75–0.87), and in the general population (206), but its benefit (relative to its risk) in primary prevention (for those without established ASCVD) is more controversial. In primary prevention, the absolute risks of vascular events are lower than in secondary prevention; however, the complication rates (i.e., bleeding) are comparable.

The Japanese Primary Prevention of Atherosclerosis with Aspirin for Diabetes (JPAD) study, initially published in 2008 after 4.4 years of follow-up, did not demonstrate a statistically significant reduction in CVD events with aspirin 81 mg or 100 mg per day among patients with type 2 diabetes but without clinical CVD (207). The 10-year follow-up data from JPAD again showed no benefit for aspirin for CVD prevention but did demonstrate an increased risk of gastrointestinal bleeding (208). Prior meta-analyses in 2009 had suggested a modest benefit of aspirin in primary prevention for CVD risk reduction among persons with and without diabetes (206,209), with potential interaction by sex (209).

However, even more recent evidence from randomized clinical trials has shown less benefit for prophylactic aspirin when used in combination with other contemporary ASCVD preventive therapies. In 2018, the ASCEND, ARRIVE, and ASPREE trials were published. The ASCEND (A Study of Cardiovascular Events in Diabetes) trial evaluated more than 15,000 adults who had diabetes but no ASCVD and found that the absolute benefit of reduction in serious vascular events by low-dose aspirin was largely counterbalanced by the increased risk of bleeding (210). The ARRIVE (Aspirin to Reduce Risk of Initial Vascular Events) trial, a primary prevention trial conducted among 12,546 adults without diabetes who were estimated to be at intermediate ASCVD risk, found no benefit of aspirin for reducing MACE but increased risk of gastrointestinal bleeding (211). Finally, in the Aspirin in Reducing Events in the Elderly (ASPREE) trial of 19,114 adults age >65 years (11% with diabetes), no reduction was seen in cardiovascular events with aspirin, but risks of bleeding and death increased (212,213).

An updated 2019 meta-analysis, which included these three new trials, found that the number needed to treat to cause a major bleeding event was actually lower than the number needed to treat to prevent an ASCVD event (210 vs. 241) in the general population, suggesting more harm than benefit (214). Additionally, another updated meta-analysis of the randomized controlled trials for aspirin in primary prevention specifically for patients with diabetes was conducted in 2020 (215). This analysis included 10 randomized controlled trials (four of which were conducted exclusively in populations with diabetes) and 34,058 patients with diabetes with median follow-up of 5.7 years. The pooled analysis showed a modest 8% reduction in MACE (RR 0.92, 95% CI 0.84–0.999) but a 30% increased risk of major bleeding (RR 1.30, 95% CI 1.10–1.53) and a lack of a mortality benefit among persons with diabetes in a primary prevention setting (215).

Trials of Comprehensive Risk Factor and Lifestyle Management

Few trials that include interventions targeted at multiple risk factors for CVD among persons with diabetes have been published (Table 7). The Steno-2 trial was conducted among 160 patients with type 2 diabetes who were randomized to intensive therapy designed to lower glucose, LDL-C, and blood pressure; after a 7.8-year follow-up, those in the intensive therapy arm had a significantly lower risk of CVD events (HR 0.47, 95% CI 0.24–0.73) (216). A further 5.5-year follow-up of this study cohort showed a subsequent 46% lower total mortality, 57% lower CVD death rate, and 59% lower risk of subsequent CVD events (217).

Table Icon

TABLE 7.

Clinical Trials of Comprehensive Risk Factor Control and Lifestyle Management on Cardiovascular Risk

A meta-analysis of clinical trials of at least 12 weeks duration in patients with type 2 diabetes evaluated the ability of structured exercise training or physical activity advice to lower A1c levels compared with a control group; 47 randomized clinical trials (n=8,538 subjects) were included (218). Structured aerobic exercise, structured resistance training, and both combined were each associated with declines in A1c levels compared with control participants.

The intensive lifestyle approach employed in the Diabetes Prevention Program (DPP) (219)—a trial in people with prediabetes, rather than diabetes—involved a targeted weight loss of 7% and 150 minutes per week of physical activity. The mean age of the 3,234 participants was 51 years; mean BMI was 34.0 kg/m2; 68% were women; 45% were members of minority groups. The lifestyle intervention compared to placebo resulted in a 58% reduction in the onset of new type 2 diabetes when the trial was stopped early based on observed benefits in preventing diabetes, with an average follow-up of 2.8 years. A study based on extended follow-up from the DPPOS (220) examined whether regression from prediabetes (defined as consistently having FPG 5.6–6.9 mmol/L [100.9–124.3 mg/dL] and/or 2-hour plasma glucose levels of 7.8–11.0 mmol/L [140.5–198.2 mg/dL] on annual OGTT during the DPP period and never having met the criteria for the diagnosis of diabetes) to normal glucose regulation has a carry-over effect in reducing long-term diabetes risk. This study in DPPOS showed a 56% reduced risk of developing diabetes over 10 years in those who returned to normal glucose during the intervention phase. However, while the intensive lifestyle intervention achieved similar benefits on blood pressure and dyslipidemia compared to metformin and placebo treatment arms in the DPPOS (221), no effect of lifestyle intervention was found on the prevention of subclinical atherosclerosis as measured by CAC compared to placebo, although there was a protective effect of metformin (222).

The largest trial to address the value of behavioral intervention for CVD prevention in adults with known diabetes is the Look AHEAD study of 5,145 U.S. adults with BMI ≥25 kg/m2 and type 2 diabetes (223). This study compared an intensive lifestyle group, consisting of weekly group and individual counseling in the first 6 months followed by three sessions for the next 6 months and refresher sessions afterwards, to diabetes support and education alone, involving three group sessions annually in the first 4 years and decreasing to one thereafter. Look AHEAD showed that those in the intensive group lost significantly more weight (6.2% vs. 0.9% of starting weight), had greater improvement in fitness, blood pressure, HDL-C, and triglycerides, and were significantly more likely to experience remission in their diabetes (223). In October 2012, the National Institutes of Health announced early termination of this trial after ≤11 years due to absent differences in CVD events and lack of a reasonable likelihood of showing a CVD difference if the study were continued for the planned 13 years (224). However, in post hoc analysis, those who lost at least 10% of their bodyweight compared to individuals with stable weight or weight gain in the first year of the Look AHEAD study had a 21% lower risk (adjusted HR 0.79, 95% CI 0.64–0.98, p=0.034) of the primary cardiovascular composite outcome, which included cardiovascular death, nonfatal acute MI, nonfatal stroke, or admission to hospital for angina (93). Other analyses from Look AHEAD (93) reported that the initial differences in benefit in risk factors and physical activity between groups diminished substantially during later years of the trial, and at the end of the 9.6-year median follow-up, the primary outcome of cardiovascular death, nonfatal MI, stroke, or hospitalization due to angina did not differ when comparing the intervention and control groups (HR 0.95, p=0.51).

Cardiovascular Outcome Trials of Glucose-Lowering Medications

Before 2008, new diabetes medications could be approved by the FDA for marketing in the United States if they lowered the surrogate endpoint A1c. These trials were often small, of short duration, and excluded patients with established CVD. However, the combination of a lack of adverse cardiovascular event reduction in trials testing glucose-lowering strategies (ACCORD, ADVANCE, VADT) and an increased mortality risk observed in ACCORD, as well as suggestion of cardiovascular harm from the rosiglitazone development program, prompted the FDA to require evidence of cardiovascular safety in new diabetes drugs (225,226). The result of this decision was that pharmaceutical companies that wanted to bring a novel antihyperglycemic medication to market in the United States were required to conduct large CVOTs in patients with established or at high risk for CVD.

Since that FDA guidance, a large number of rigorously conducted cardiovascular and kidney safety and efficacy trials have been conducted. Many more are planned or underway (227). The investment in this area of endocrine, cardiovascular, and renal medicine speaks to the clinical importance of the area, as well as to the unprecedented cardiovascular event reduction seen with two novel classes of medications originally designed and marketed as glucose reduction agents: the sodium-glucose cotransporter-2 inhibitors (SGLT2i) and the glucagon-like peptide-1 receptor agonists (GLP-1 RA). While this section focuses on SGLT2i and GLP-1 RAs, large outcome trials have been conducted for dipeptidyl peptidase-4 inhibitors (DPP4i), long-acting insulin (insulin glargine and insulin degludec), a thiazolidinedione, and alpha-glucosidase inhibitors (227). Furthermore, trials testing dual SGLT2 and SGLT1 inhibitors have been published (228,229), and the dual GLP-1 RA and glucose-dependent insulinotropic polypeptide (GIP) agonist, tirzepatide, approved in 2022 by the FDA for treatment of type 2 diabetes, is being studied in ongoing CVOTs (230).

Sodium-Glucose Cotransporter-2 Inhibitors

SGLT2i reduce glucose reabsorption in the proximal tubule of the kidney, thereby leading to reductions in A1c through glucosuria. The SGLT2i that have demonstrated benefit in reducing MACE among people with diabetes are empagliflozin, canagliflozin, and dapagliflozin. The SGLT1/2i sotagliflozin also has demonstrated cardiovascular benefit in people with diabetes. The trials in which SGLT2i medications were tested and their outcomes are listed in Table 8 (228,229,231,232,233,234). A meta-analysis reported an 11% reduction in the risk of 3-point MACE, a 23% reduction in the risk of hospitalization for HF or cardiovascular mortality, and a 16% reduction in all-cause mortality with SGLTi (235). The mechanism of the effects of SGLT2i on ASCVD, HF, and cardiovascular death outcomes are not well established but do appear to be largely independent of their effects on blood glucose. Indeed, in trials in patients with HF who were not required to have type 2 diabetes for eligibility, dapagliflozin and empagliflozin showed substantial benefit in patients with HF with reduced ejection fraction (236,237) and in patients with HF with preserved ejection fraction (238,239). SGLT2i also have important benefits for patients with chronic kidney disease (CKD) with or without diabetes, as discussed further in the SGLT2 Inhibitors in Chronic Kidney Disease section; a meta-analysis of published trials estimated a 45% reduction in the risk of worsening estimated glomerular filtration rate (eGFR), end-stage kidney disease, or kidney death (HR 0.55, 95% CI 0.48–0.64) in those randomized to active SGLT2i therapy (235).

Table Icon

TABLE 8.

Cardiovascular Outcome Trials of SGLT2 Inhibitors in Patients With Type 2 Diabetes

Glucagon-Like Peptide-1 Receptor Agonists

GLP-1 RAs mimic the action of endogenous GLP-1 by mimicking glucose-dependent insulin secretion and inhibiting glucagon secretion from the pancreas. The effects of GLP-1 RAs on cardiovascular outcomes are heterogeneous, with some agents (liraglutide, semaglutide SQ, dulaglutide, albiglutide, and efpeglenatide) demonstrating clinically important reduction in MACE (Table 9) (240,241,242,243,244,245,246,247). Other agents, including lixisenatide and exenatide, have demonstrated cardiovascular safety but no evidence of cardiovascular benefit (240,243). The oral formulation of semaglutide has shown evidence of cardiovascular safety, but the published trial (PIONEER-6) was not powered to demonstrate cardiovascular benefit (246). A larger CVOT with oral semaglutide is ongoing (the SOUL trial, NCT03914326). A meta-analysis of randomized trials of GLP-1 RAs reported a 12% reduction in MACE, 12% reduction in cardiovascular mortality, 16% reduction in the risk of fatal and nonfatal stroke, and 9% reduction in the risk of fatal and nonfatal MI (248). The authors also noted a 12% reduction in all-cause mortality and 9% reduction in hospitalization for HF; all reductions were statistically significant. The mechanisms by which GLP-1 RAs exert their cardiovascular benefit are unknown but are at best only partially explained by their effects on blood glucose (A1c reduction). GLP-1 RAs may exert their cardiovascular benefit through beneficial changes in cardiovascular risk factors, including blood pressure, lipids, weight, or subclinical inflammation (249).

Table Icon

TABLE 9.

Cardiovascular Outcome Trials of GLP-1 RA in Patients With Type 2 Diabetes

Unanswered Questions for Patients and Providers

As described, ample evidence is available from placebo-controlled CVOTs that both SGLT2i and GLP-1 RAs reduce MACE, hospitalization for HF, and other clinically important outcomes for patients with type 2 diabetes. However, a number of key clinical questions have yet to be answered, including whether the medications are as effective at preventing MACE in lower-risk (i.e., without established CVD) patients; whether one class of therapy is better than the other for the prevention of cardiovascular outcomes and, if so, for which outcomes in which patients; and whether combination therapy with SGLT2i and GLP-1 RAs will offer benefits that are additive, more than additive, or sub-additive.

Some of these questions have begun to be addressed with the inclusion of lower-risk patients in randomized trials. In those analyses, summarized in a meta-analysis, SGLT2i do not appear to reduce atherosclerotic cardiovascular events like MI or stroke in patients without established CVD. However, these drugs have substantial beneficial effects on HF hospitalization and progression of kidney disease in patients with and without ASCVD at baseline (250). In a meta-analysis of randomized trials of GLP-1 RAs, the effect on cardiovascular outcomes appeared similar across the presence or absence of CVD at baseline (249). Observational pharmacoepidemiologic studies have compared SGLT2i to GLP-1 RAs and raised the possibility that SGLT2i may be superior to GLP-1 RAs among patients with established CVD in hospitalization for HF (251). Large comparative effectiveness interventional trials have been planned and are underway to address which therapy is better for which outcomes in which patients and to compare combination SGLT2i and GLP-1 RA therapy to monotherapy with either agent (252).

Cardiovascular Outcomes in Diabetic Kidney Disease and a Role for Novel Therapeutic Agents

CKD is highly prevalent among individuals with diabetes (~40%) (253), and the presence of diabetic kidney disease is associated with a substantially increased risk of CVD. CVD is the leading cause of death in the diabetic kidney disease population (254). Furthermore, reduction in eGFR and the presence of albuminuria are independently associated with increased risks for CVD and mortality in a graded fashion (255,256).

CKD among persons with diabetes can contribute to worse cardiovascular outcomes through several mechanisms, such as progression of atherosclerotic disease, valvular calcification, and myocardial fibrosis. Patients with CKD are more likely to die from cardiovascular causes than from progression to end-stage kidney disease. Accordingly, CKD is considered a risk-enhancing factor in persons with diabetes in the ACC/AHA primary prevention guideline (257). The risk of cardiovascular mortality is greater in the presence of having both diabetes and CKD (16%, 95% CI 11%–21%) compared to the risks conferred by diabetes alone (3%) or CKD alone (6%) (258). Thus, persons who have both diabetes and CKD need even more intensive preventive strategies.

The first-line therapy for diabetic kidney disease is treatment with angiotensin-converting enzyme inhibitors (ACEI) or angiotensin receptor blockers (ARB), which have both kidney and cardioprotective properties (254,259). As mentioned, newer classes of medications are now available that reduce both CKD progression and cardiovascular events among persons with diabetic kidney disease, which include the SGLT2i and GLP-1 RA agents, as well as the nonsteroidal mineralocorticoid receptor antagonist (MRA) finerenone.

SGLT2 Inhibitors in Chronic Kidney Disease

SGLT2i significantly reduce cardiovascular and kidney outcomes in patients with type 2 diabetes, CKD, and/or HF (260), as described above. In two trials enrolling exclusively a CKD population, the CREDENCE (Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation) (261) and the DAPA-CKD (Dapagliflozin and Prevention of Adverse Outcomes in Chronic Kidney Disease) (262) trials, the SGLT2 inhibitors canagliflozin and dapagliflozin, respectively, reduced the risk for both the primary adverse kidney outcomes and also the secondary cardiovascular outcomes among patients with CKD.

The CREDENCE trial enrolled patients with albuminuric diabetic kidney disease with eGFR ≥30 mL/min/1.73 m2 and showed that the canagliflozin-treated patients had 20% lower risk of cardiovascular death, MI, or stroke (HR 0.80, 95% CI 0.67–0.95) and 39% lower risk of hospitalization for HF (HR 0.61, 95% CI 0.47–0.90) compared to placebo (261).

In the DAPA-CKD trial, which enrolled CKD patients with or without diabetes with eGFR ≥25 mL/min/1.73 m2, dapagliflozin conferred a 29% reduction in HF hospitalization and cardiovascular death (HR 0.71, 95% CI 0.55–0.92) with similar benefit regardless of diabetes status (262).

The SCORED trial, which also enrolled an exclusive CKD population, demonstrated cardiovascular risk reduction with sotagliflozin (228).

Another dedicated trial in an exclusive CKD population evaluated empagliflozin in the broadest CKD population to date, including those with and without diabetes and with and without albuminuria (EMPA-KIDNEY). The trial was stopped early for clear efficacy of empagliflozin in reducing the progression of kidney disease or death from cardiovascular causes compared to placebo (263). The results of this trial are informative about the benefits of SGLT2i among various CKD subgroups, including nonalbuminuric CKD.

GLP-1 Receptor Agonists in Chronic Kidney Disease

As mentioned, the long-acting injectable GLP-1 RAs reduce ASCVD events in patients with type 2 diabetes, with similar benefit for patients with and without CKD (249,254,264). In a meta-analysis of GLP-1 RA randomized clinical trials, the reduction in 3-point MACE (cardiovascular death, MI, and stroke) was 15% overall, with hazard ratios of 0.88 (95% CI 0.77–1.01) and 0.83 (95% CI 0.74–0.93) for eGFR <60 and ≥60 mL/min/1.73 m2, respectively (p-interaction=0.51). GLP-1 RAs also reduce kidney outcomes, primarily driven by reduction in albuminuria (249). A dedicated kidney outcome trial for a GLP-1 RA (semaglutide in the FLOW trial, NCT03819153) was stopped early in October 2023 due to clear efficacy, with results anticipated to read out in 2024.

Mineralocorticoid Receptor Antagonist in Chronic Kidney Disease

For patients with diabetic kidney disease, the nonsteroidal MRA finerenone was demonstrated to reduce cardiovascular events, as well as to slow the progression of kidney disease, in two large outcome trials (265,266). The FIDELITY analysis pooled the data from the FIDELIO-DKD and the FIGARO-DKD trials, including more than 13,000 individuals with type 2 diabetes and CKD followed for a median of 3.0 years (267). These data showed that finerenone reduced the composite cardiovascular outcome (cardiovascular death, nonfatal MI, nonfatal stroke, or HF hospitalization) by 14% (HR 0.86, 95% CI 0.78–0.95), in addition to significantly reducing the composite kidney outcome by 23%. Finerenone was associated with a small increased risk of hyperkalemia requiring treatment discontinuation (1.7% vs. 0.6% in placebo). Notably, only individuals with a serum potassium level ≤4.8 mmol/L were enrolled in these finerenone outcome studies, and safety was ensured by close monitoring of serum potassium levels, with discontinuation of study drug if potassium exceeded 5.5 mmol/L (could be restarted if potassium decreased to below 5 mmol/L).

Evidence-Based Recommendations for Diabetes Management From Professional Societies

Many professional societies have recommendations for pharmacologic therapy for cardiovascular risk factor and CVD prevention in people with type 2 diabetes (75,268,269,270) in conjunction with lifestyle modifications, which include ≥5% weight loss through individualized medical nutrition (diet), physical activity, and/or behavioral therapy. These recommendations are summarized in Table 10 and detailed below.

Table Icon

TABLE 10.

Evidence-Based Clinical Recommendations From Professional Societies for the Pharmacological Prevention of Cardiovascular Disease in Patients with Diabetes, 2023

Clinical Guidelines for CVD Prevention in People With Diabetes

Landmark trials of intensive versus standard glucose-lowering strategies have demonstrated significant reduction in microvascular complications, but uncertain benefits for CVD, and inform current clinical practice. According to the ADA recommendations in 2023, most nonpregnant adults may benefit from an A1c target of <7%. However, individualizing A1c goals—so as to avoid harm (as was seen in ACCORD)—and additionally optimizing multiple cardiovascular risk factors beyond glucose, while still striving for tight glycemic control in those for whom it can be reasonably and safely achieved, are now major tenets in the care of people with diabetes (75,271).

All patients across the risk spectrum benefit from the implementation of favorable lifestyle changes, including regular physical activity and healthy diet patterns, to improve their glucose, blood pressure, and lipid parameters. Major professional societies recommend treating with antihypertensive drug therapy to a blood pressure target of <130/80 mmHg in patients with type 2 diabetes and hypertension (74,75). The ADA and ACC/AHA guidelines recommend at least a moderate-intensity statin for individuals age >40 years who have diabetes for primary prevention of ASCVD (75,179,257).

However, there may be situations of statin intolerance or patient preference to avoid pharmacotherapy where additional risk stratification may be desired. Some individuals with diabetes are actually at lower ASCVD risk than might be predicted given their diabetes status, and the CAC scores can be useful in selecting patients with diabetes to reclassify risk and identify lower and high-risk individuals, when risk status is otherwise uncertain (272,273). One caveat with the use of CAC as a risk marker is that it takes some time for plaque to calcify, and the absence of CAC may be less reassuring in younger compared to older adults. In younger patients age <40 years with diabetes, a CAC score of zero may not sufficiently identify low-risk individuals. The presence of non-calcified plaque can be identified in younger individuals with diabetes by contrast CT angiography (CCTA) (274), but at this time, CCTA is not recommended solely for the purpose of screening among asymptomatic individuals. However, nonobstructive coronary artery disease is not benign and is associated with increased risk of vascular events (275,276). Thus, if an individual undergoes CCTA for the purpose of evaluation of chest pain and is found to have nonobstructive plaque, intensified preventive therapy with statins and/or other LDL-C-lowering therapies as necessary would be recommended to slow atherosclerosis progression and reduce risk of ASCVD events (277,278).

Patients with diabetes also have greater risk of carotid plaque compared to their counterparts without diabetes (279). While current guidelines do not endorse cIMT for screening, the presence of carotid plaque identified by CT or ultrasound is a marker of atherosclerosis and predictive of ASCVD risk, including among patients with type 2 diabetes (280). ABI testing has low sensitivity for identifying individuals at risk and, thus, is not endorsed for a routine screening test. If performed, the presence of a low ABI (<0.9) is a risk-enhancing factor, particularly among individuals with diabetes, that would favor more intensified preventive therapy, such as use of high-intensity statin (179,257).

Trial findings have also guided updated aspirin recommendations in the 2019 ACC/AHA guideline for the primary prevention of CVD (257). These recommendations differed from prior AHA guidelines that had recommended aspirin for patients with 10-year ASCVD risk ≥10%. The 2019 ACC/AHA guidelines state that most healthy people do not need to take aspirin. However, aspirin could be considered for select patients age 40–70 years who have a very high risk of ASCVD and may benefit from aspirin if at low risk for bleeding (IIb recommendation). However, these decisions are needed in the context of a clinician-patient risk discussion. In 2022, the U.S. Preventive Services Task Force (USPSTF) also published updated recommendations for the use of aspirin in primary prevention, which downgraded their prior aspirin recommendations in their 2016 statement (281). In their revised recommendations, aspirin in primary prevention may be considered selectively for people age 40–59 years who are at higher risk for CVD (>10% 10-year risk) (C-grade recommendation). However, persons age ≥60 years should not start taking aspirin for primary CVD prevention (D grade recommendation). The USPSTF further acknowledges that relative bleeding risk linked with aspirin is consistent by age, sex, diabetes status, and level of CVD risk, but absolute bleeding risk is higher among older adults (281). In contrast, in 2023, the ADA still gave a level A recommendation that 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, but aspirin therapy is not generally recommended in those age >70 years (75).

In patients with established ASCVD or at high risk of ASCVD, numerous professional societies recommend that the regimen include a GLP-1 RA and/or SGLT2i with demonstrated CVD benefit to reduce cardiovascular events independent of baseline A1c, individualized A1c target, or metformin use. In those with HF who have either reduced or preserved ejection fraction, inclusion of an SGLT2i with demonstrated benefit is recommended to reduce the risk of HF hospitalization and cardiovascular death (75,268,269,270).

Among high-risk patients with diabetic kidney disease, ACEI/ARB, SGLT2i, GLP-1 RA, and finerenone have all been demonstrated to significantly reduce the risk of adverse cardiovascular outcomes. Since the trials of the latter three agents were conducted on the background of ACEI or ARB, ACEI/ARB should be initiated first if no contraindications. Both cardiology and nephrology guidelines strongly endorse the use of SGLT2i for cardiovascular and kidney protection for patients with diabetic kidney disease; so, SGLT2i should be prioritized next in this population (254,270). GLP-1 RAs may be additionally recommended, particularly if further A1c lowering is needed. Future guidelines will likely address how best to implement finerenone in treatment algorithms; finerenone was approved by the FDA in 2021. Patient characteristics, drug availability, and cost considerations likely will influence choices as part of shared decision-making. The ADA recommends measuring both eGFR and the urine albumin-creatinine ratio (UACR) annually in patients with diabetes; however, this is suboptimally done in clinical practice with approximately half of patients with diabetes receiving both laboratory tests (282). Unfortunately, many patients with diabetic kidney disease, who are at elevated cardiovascular risk, go unrecognized without an intentional assessment for albuminuria.

Risk Assessment and Stratification

Primary Prevention

When considering drug therapy, estimation of ASCVD risk facilitates matching intensity of therapy to a patient’s absolute risk of having a cardiovascular event to maximize anticipated benefits of therapy and minimize harms of over-treatment. The 2019 ACC/AHA Guideline for the Primary Prevention of CVD still gives a class I recommendation that for adults age 40–74 years without known CVD, clinicians should routinely assess traditional cardiovascular risk factors and estimate 10-year risk of ASCVD using the pooled cohort equations (PCE) (34,257). The PCE estimates 10-year risk of a hard ASCVD event, which means MI, CHD death, or stroke; they are race- and sex-specific equations that are best calibrated for non-Hispanic White and Black adults living in the United States (34). The input variables are the same as for the older Framingham Risk Equation and include age, sex, race, total cholesterol, HDL-C, systolic blood pressure, treatment for hypertension, diabetes, and smoking.

The 2019 ACC/AHA guideline acknowledges limitations of the PCE, which can over- or under-estimate ASCVD risk in certain populations. Therefore, the next recommended step is to consider risk-enhancing factors, which are clinical or laboratory factors not included in the PCE that are also associated with elevated ASCVD risk. These factors include having a family history of premature ASCVD, metabolic syndrome, CKD, the female-specific factors of early menopause or preeclampsia, South Asian ancestry, chronic inflammatory conditions, such as lupus or rheumatoid arthritis, persistently elevated triglycerides, elevated hsCRP, elevated apolipoprotein B, elevated lipoprotein (a), or low ABI (257). The presence of risk-enhancing factors would favor the initiation of statin therapy in individuals who are otherwise at borderline or intermediate estimated risk.

There are some important caveats for persons with diabetes. While diabetes is a factor included in the PCE, it is not really meant to be applied to persons with diabetes when deciding on initiation of statin therapy. The 2019 ACC/AHA guidelines recommend that for adults age 40–75 years who have diabetes, regardless of their estimated 10-year ASCVD risk, a moderate-intensity statin is indicated for prevention (class I recommendation). Furthermore, for persons with diabetes with multiple ASCVD risk factors, it is reasonable to prescribe a high-intensity statin with the aim of lowering LDL-C by 50% or more (class IIa recommendation) (257). In addition to the traditional ASCVD factors, the ACC/AHA guidelines also highlight diabetes-specific risk-enhancing factors, which include long duration of diabetes (≥10 years for type 2 diabetes and ≥20 years for type 1 diabetes), albuminuria ≥30 mcg albumin/mg creatinine, eGFR <60 mL/min/1.73 m2, retinopathy, neuropathy, or ABI <0.9 (257). The presence of these factors would also favor initiation of a high-intensity statin.

The ADA Standards of Care 2023 similarly endorse that cardiovascular risk factors should be assessed at least annually in patients with diabetes, including duration of diabetes, overweight/obesity, hypertension, dyslipidemia, smoking, family history of premature CHD, CKD, and presence of albuminuria (75). The ADA encourages the use of the PCE for 10-year risk assessment as a useful starting tool.

While risk factor assessment and treatment are strongly recommended, the ACC/AHA and the ADA do not recommend routine screening for coronary artery disease via stress testing in asymptomatic individuals as it does not improve outcomes as long as cardiovascular risk factors are optimally treated (75,257). Selective stress testing or other cardiovascular imaging (echocardiography, etc.) can be considered for patients with symptoms concerning for CVD or who have other abnormal physical exam findings or electrocardiogram (ECG) evidence suggestive of ASCVD or HF.

Although younger individuals age <40 years are not included in the 10-year risk assessment, it is still reasonable to routinely assess traditional ASCVD risk factors. For adults age 20–39 years or for those age 40–59 years who are at low (<7.5%) 10-year ASCVD risk, clinicians may consider estimating a lifetime or 30-year ASCVD risk score (also available by the PCE) to help facilitate lifestyle changes and monitoring of risk factors (257).

Secondary Prevention

All patients age <75 years with established ASCVD are recommended for a high-intensity statin (with those age ≥75 years considered for a moderate- or high-intensity statin). However, the guidelines acknowledge that even within a secondary prevention population, risk for a subsequent event is not heterogeneous. Therefore, the 2018 AHA/ACC/Multi-Society Cholesterol Guideline further subdivides their recommendations into ASCVD “Not at Very High Risk” and ASCVD “At Very High Risk,” with differing recommendations about the intensity of LDL-C lowering (179).

Patients at very high risk include those with a major ASCVD event, such as recent ACS, prior MI, ischemic stroke, or symptomatic PAD, with at least one other high-risk condition, including age ≥65 years, CKD, hypertension, current smoking, persistently elevated LDL-C, and diabetes (179). Therefore, the presence of diabetes in the setting of a recent ACS, MI, PAD, or stroke elevates patients into this very high-risk category that warrants the most intensive preventive intervention. The 2019 AHA/ACC guidelines recommend the addition of a PCSK9i for high-risk secondary prevention patients, if LDL-C remains above a threshold of ≥70 mg/dL despite statin and ezetimibe therapy or above a threshold ≥100 mg/dL (≥2.59 mmol/L) for patients with familial hypercholesterolemia in primary prevention. Recent guidelines further recommend that patients with ASCVD at “very high risk” achieve an LDL-C threshold <55 mg/dL.

Conclusion

Among people with diabetes in the United States, the prevalence of CVD remains overall about twofold greater than in people without diabetes, although sex differences also exist. These disparities represent a major health care challenge.

The relevance of dysglycemia to risk of CVD remains uncertain in people with prediabetes or diabetes, which raises questions about the role of hyperglycemia per se. Intensive versus standard glucose-lowering strategies and mortality have not necessarily demonstrated CVD benefit, and instead, some clinical trial data suggest that intensive management of moderate hyperglycemia may cause harm in high-risk subjects. Also, despite the increase in diabetes prevalence, little evidence exists that measures of obesity are related to CVD independent of other risk factors or that weight reduction through change in lifestyle yields benefit for CVD prevention. Traditional risk factors, such as dyslipidemia, hypertension, and smoking, clearly are important, although some studies call into question the importance of mild elevations in blood pressure in diabetes.

Pharmacological options for the primary and secondary prevention of CVD include the use of statin therapy, which is widely recommended for most people with diabetes age ≥40 years. Additionally, ezetimibe and PCSK9i have cardiovascular benefit among high-risk individuals with residual LDL-C elevation on maximally tolerated statin treatment, while bempedoic acid was demonstrated to have cardiovascular benefit in patients who are intolerant to statin therapy. However, despite the importance of elevated triglycerides and low HDL-C as risk factors, the cardiovascular outcome benefit of targeting lipid fractions beyond LDL-C is unproven. IPE has CVD benefit among individuals with elevated triglyceride levels despite the use of statins, although the mechanism is independent of triglyceride-lowering. To date, no trials have conclusively demonstrated a benefit of fibrate therapy when added to statin therapy among persons with diabetes. A large trial in people with diabetes has demonstrated benefit of aspirin in reducing vascular events, but this benefit was counterbalanced by an increased risk of major bleeding.

The use of newer glucose-lowering agents to reduce risk of major cardiovascular events and cardiovascular deaths has been demonstrated over recent years among those with a history of established CVD and also for those at high risk, such as those with CKD. In addition, SGLT2i have demonstrated benefit in reducing hospitalization for HF and reducing CKD progression.

An improved understanding of the natural history of atherosclerosis in diabetes and its risk factors during the earlier phases of diabetes development will continue to facilitate the development of innovative and targeted therapies to prevent CVD in the future.

List of Abbreviations

A1c

glycosylated or glycated hemoglobin

ABI

ankle-brachial index

ACC

American College of Cardiology

ACCORD

Action to Control Cardiovascular Risk in Diabetes

ACEI

angiotensin-converting enzyme inhibitor

ACS

acute coronary syndrome

ADA

American Diabetes Association

ADVANCE

Action in Diabetes and Vascular Disease: Preterax and Diamicron Evaluation

AHA

American Heart Association

ARB

angiotensin receptor blocker

ARIC

Atherosclerosis Risk in Communities study

ASCVD

atherosclerotic cardiovascular disease

BMI

body mass index

BNP

B-type natriuretic peptides

BRFSS

Behavioral Risk Factor Surveillance System

CAC

coronary artery calcium

CCTA

contrast CT angiography

CDC

Centers for Disease Control and Prevention

CHD

coronary heart disease

CI

confidence interval

cIMT

carotid intimal medial thickness

CKD

chronic kidney disease

CLEAR

Cholesterol Lowering via Bempedoic Acid, an ACL-Inhibiting Regimen trial

CREDENCE

Canagliflozin and Renal Events in Diabetes with Established Nephropathy Clinical Evaluation trial

CRP

C-reactive protein

CT

computed tomography

CVD

cardiovascular disease

CVOT

cardiovascular outcome trial

DAPA-CKD

Dapagliflozin and Prevention of Adverse Outcomes in Chronic Kidney Disease trial

DCCT

Diabetes Control and Complications Trial

DHA

docosahexaenoic acid

DPP

Diabetes Prevention Program

DPPOS

Diabetes Prevention Program Outcomes Study

EDIC

Epidemiology of Diabetes Interventions and Complications study

eGFR

estimated glomerular filtration rate

EPA

eicosapentaenoic acid

FDA

U.S. Food and Drug Administration

FIELD

Fenofibrate Intervention and Event Lowering in Diabetes trial

FOURIER

Further Cardiovascular Outcomes Research With PCSK9 Inhibition in Subjects With Elevated Risk trial

FPG

fasting plasma glucose

GLP-1

glucagon-like peptide-1

GLP-1 RA

glucagon-like peptide-1 receptor agonist

HDL-C

high-density lipoprotein-cholesterol

HF

heart failure

HR

hazard ratio

hsCRP

high-sensitivity C-reactive protein

hs-cTn

high-sensitivity cardiac troponin

IMPROVE IT

Improved Reduction of Outcomes: Vytorin Efficacy International Trial

IPE

icosapent ethyl

JPAD

Japanese Primary Prevention of Atherosclerosis with Aspirin for Diabetes study

LDL-C

low-density lipoprotein-cholesterol

Look AHEAD

Action for Health in Diabetes

MACE

major adverse cardiovascular event

MESA

Multi-Ethnic Study of Atherosclerosis

MI

myocardial infarction

MRA

mineralocorticoid receptor antagonist

MRFIT

Multiple Risk Factor Intervention Trial

NHANES

National Health and Nutrition Examination Survey

NHIS

National Health Interview Survey

NT-proBNP

N-terminal proB-type natriuretic peptides

OGTT

oral glucose tolerance test

OR

odds ratio

PAD

peripheral artery disease

PCE

pooled cohort equations

PCSK9i

proprotein convertase subtilisin/kexin type 9 inhibitors

REDUCE-IT

Reduction of Cardiovascular Events with Icosapent Ethyl–Intervention Trial

RR

relative risk

SD

standard deviation

SGLT2i

sodium-glucose cotransporter-2 inhibitor

SPRINT

Systolic Blood Pressure Intervention Trial

UKPDS

United Kingdom Prospective Diabetes Study

USPSTF

U.S. Preventive Services Task Force

VADT

Veterans Affairs Diabetes Trial

WHO

World Health Organization

Conversions

A1c: (% x 10.93) - 23.50 = mmol/mol

Glucose: mg/dL x 0.0555 = mmol/L

HDL-C or LDL-C: mg/dL x 0.0259 = mmol/L

Triglycerides: mg/dL x 0.0113 = mmol/L

Acknowledgment

This is an update of: Barrett-Connor E, Wingard D, Wong N, Goldberg R: Heart Disease and Diabetes. Chapter 18 in Diabetes in America, 3rd ed. Cowie CC, Casagrande SS, Menke A, Cissell MA, Eberhardt MS, Meigs JB, Gregg EW, Knowler WC, Barrett-Connor E, Becker DJ, Brancati FL, Boyko EJ, Herman WH, Howard BV, Narayan KMV, Rewers M, Fradkin JE, Eds. Bethesda, MD, National Institutes of Health, NIH Pub No. 17-1468, 2018, p. 18.1–18.30

Article History

Received in final form on January 5, 2023.

References

1.
West KM: Epidemiology of Diabetes and Its Vascular Lesions. New York, NY, Elsevier, 1978
2.
WHO Expert Committee on Diabetes Mellitus: second report. World Health Organ Tech Rep Ser 646:1–80, 1980 [PubMed: 6771926]
3.
Classification and diagnosis of diabetes mellitus and other categories of glucose intolerance. National Diabetes Data Group. Diabetes 28:1039–1057, 1979 [PubMed: 510803]
4.
Centers for Disease Control and Prevention. Diabetes Fast Facts [article online], 2023. Available from https://www​.cdc.gov/diabetes​/basics/quick-facts.html. Accessed 9 Oct 2023
5.
Skyler JS, Sosenko JM: The evolution of type 1 diabetes. JAMA 309:2491–2492, 2013 [PubMed: 23780463]
6.
Laing SP, Swerdlow AJ, Slater SD, Burden AC, Morris A, Waugh NR, Gatling W, Bingley PJ, Patterson CC: Mortality from heart disease in a cohort of 23,000 patients with insulin-treated diabetes. Diabetologia 46:760–765, 2003 [PubMed: 12774166]
7.
Kalyani RR: Glucose-lowering drugs to reduce cardiovascular risk in type 2 diabetes. N Engl J Med 384:1248–1260, 2021 [PubMed: 33789013]
8.
Barrett-Connor E, Wingard D, Wong N, Goldberg R: Heart Disease and Diabetes. Chapter 18 in Diabetes in America, 3rd ed. Cowie CC, Casagrande SS, Menke A, Cissell MA, Eberhardt MS, Meigs JB, Gregg EW, Knowler WC, Barrett-Connor E, Becker DJ, Brancati FL, Boyko EJ, Herman WH, Howard BV, Narayan KMV, Rewers M, Fradkin JE, Eds. Bethesda, MD, National Institutes of Health, NIH Pub No. 17-1468, 2018, p. 18.1–18.30. Available from https://www​.ncbi.nlm​.nih.gov/books/NBK568001/
9.
Schramm TK, Gislason GH, Kober L, Rasmussen S, Rasmussen JN, Abildstrom SZ, Hansen ML, Folke F, Buch P, Madsen M, Vaag A, Torp-Pedersen C: Diabetes patients requiring glucose-lowering therapy and nondiabetics with a prior myocardial infarction carry the same cardiovascular risk: a population study of 3.3 million people. Circulation 117:1945–1954, 2008 [PubMed: 18378618]
10.
Spencer EA, Pirie KL, Stevens RJ, Beral V, Brown A, Liu B, Green J, Reeves GK; Million Women Study Collaborators: Diabetes and modifiable risk factors for cardiovascular disease: the prospective Million Women Study. Eur J Epidemiol 23:793–799, 2008 [PubMed: 19015938]
11.
Huxley R, Barzi F, Woodward M: Excess risk of fatal coronary heart disease associated with diabetes in men and women: meta-analysis of 37 prospective cohort studies. BMJ 332:73–78, 2006 [PMC free article: PMC1326926] [PubMed: 16371403]
12.
Woodward M, Zhang X, Barzi F, Pan W, Ueshima H, Rodgers A, MacMahon S; Asia Pacific Cohort Studies Collaboration: The effects of diabetes on the risks of major cardiovascular diseases and death in the Asia-Pacific region. Diabetes Care 26:360–366, 2003 [PubMed: 12547863]
13.
Beckman JA, Creager MA, Libby P: Diabetes and atherosclerosis: epidemiology, pathophysiology, and management. JAMA 287:2570–2581, 2002 [PubMed: 12020339]
14.
Coutinho M, Gerstein HC, Wang Y, Yusuf S: The relationship between glucose and incident cardiovascular events. A metaregression analysis of published data from 20 studies of 95,783 individuals followed for 12.4 years. Diabetes Care 22:233–240, 1999 [PubMed: 10333939]
15.
Lawes CMM, Parag V, Bennett DA, Suh I, Lam TH, Whitlock G, Barzi F, Woodward M; Asia Pacific Cohort Studies Collaboration: Blood glucose and risk of cardiovascular disease in the Asia Pacific region. Diabetes Care 27:2836–2842, 2004 [PubMed: 15562194]
16.
Helfand M, Buckley DI, Freeman M, Fu R, Rogers K, Fleming C, Humphrey LL: Emerging risk factors for coronary heart disease: a summary of systematic reviews conducted for the U.S. Preventive Services Task Force. Ann Intern Med 151:496–507, 2009 [PubMed: 19805772]
17.
Emerging Risk Factors Collaboration; Sarwar N, Gao P, Seshasai SR, Gobin R, Kaptoge S, Di Angelantonio E, Ingelsson E, Lawlor DA, Selvin E, Stampfer M, Stehouwer CD, Lewington S, Pennells L, Thompson A, Sattar N, White IR, Ray KK, Danesh J: Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: a collaborative meta-analysis of 102 prospective studies. Lancet 375:2215–2222, 2010 [PMC free article: PMC2904878] [PubMed: 20609967]
18.
Centers for Disease Control and Prevention. Adult Obesity Facts [article online], 2022. Available from https://www​.cdc.gov/obesity/data/adult​.html.
19.
Centers for Disease Control and Prevention. National Diabetes Statistics Report: Appendix [article online], 2022. Available from https://www​.cdc.gov/diabetes​/data/statistics-report​/appendix.html#print. Accessed 10 Oct 2022
20.
Kalyani RR, Lazo M, Ouyang P, Turkbey E, Chevalier K, Brancati F, Becker D, Vaidya D: Sex differences in diabetes and risk of incident coronary artery disease in healthy young and middle-aged adults. Diabetes Care 37:830–838, 2014 [PMC free article: PMC3931379] [PubMed: 24178997]
21.
Gillum RF, Mussolino ME, Madans JH: Diabetes mellitus, coronary heart disease incidence, and death from all causes in African American and European American women: the NHANES I epidemiologic follow-up study. J Clin Epidemiol 53:511–518, 2000 [PubMed: 10812324]
22.
Volgman AS, Palaniappan LS, Aggarwal NT, Gupta M, Khandelwal A, Krishnan AV, Lichtman JH, Mehta LS, Patel HN, Shah KS, Shah SH, Watson KE; American Heart Association Council on Epidemiology and Prevention; Cardiovascular Disease and Stroke in Women and Special Populations Committee of the Council on Clinical Cardiology; Council on Cardiovascular and Stroke Nursing; Council on Quality of Care and Outcomes Research; Stroke Council: Atherosclerotic cardiovascular disease in South Asians in the United States: epidemiology, risk factors, and treatments: a Scientific Statement from the American Heart Association. Circulation 138:e1–e34, 2018 [PubMed: 29794080]
23.
Tanasescu M, Leitzmann MF, Rimm EB, Hu FB: Physical activity in relation to cardiovascular disease and total mortality among men with type 2 diabetes. Circulation 107:2435–2439, 2003 [PubMed: 12719277]
24.
Broekhuizen LN, Boekholdt SM, Arsenault BJ, Despres JP, Stroes ES, Kastelein JJ, Khaw KT, Wareham NJ: Physical activity, metabolic syndrome, and coronary risk: the EPIC-Norfolk prospective population study. Eur J Cardiovasc Prev Rehabil 18:209–217, 2011 [PubMed: 21450666]
25.
Yudkin JS: How can we best prolong life? Benefits of coronary risk factor reduction in non-diabetic and diabetic subjects. BMJ 306:1313–1318, 1993 [PMC free article: PMC1677709] [PubMed: 8518573]
26.
Ockene JK, Kuller LH, Svendsen KH, Meilahn E: The relationship of smoking cessation to coronary heart disease and lung cancer in the Multiple Risk Factor Intervention Trial (MRFIT). Am J Public Health 80:954–958, 1990 [PMC free article: PMC1404774] [PubMed: 2368857]
27.
Clair C, Rigotti NA, Porneala B, Fox CS, D’Agostino RB, Pencina MJ, Meigs JB: Association of smoking cessation and weight change with cardiovascular disease among adults with and without diabetes. JAMA 309:1014–1021, 2013 [PMC free article: PMC3791107] [PubMed: 23483176]
28.
Marmot M, Friel S, Bell R, Houweling TA, Taylor S; Commission on Social Determinants of Health: Closing the gap in a generation: health equity through action on the social determinants of health. Lancet 372:1661–1669, 2008 [PubMed: 18994664]
29.
Havranek EP, Mujahid MS, Barr DA, Blair IV, Cohen MS, Cruz-Flores S, Davey-Smith G, Dennison-Himmelfarb CR, Lauer MS, Lockwood DW, Rosal M, Yancy CW; American Heart Association Council on Quality of Care and Outcomes Research; Council on Epidemiology and Prevention; Council on Cardiovascular and Stroke Nursing; Council on Lifestyle and Cardiometabolic Health; Stroke Council: Social determinants of risk and outcomes for cardiovascular disease: a Scientific Statement from the American Heart Association. Circulation 132:873–898, 2015 [PubMed: 26240271]
30.
Beckles GL, Chou CF: Disparities in the prevalence of diagnosed diabetes—United States, 1999–2002 and 2011–2014. MMWR Morb Mortal Wkly Rep 65:1265–1269, 2016 [PubMed: 27855140]
31.
Gaskin DJ, Thorpe RJ, Jr., McGinty EE, Bower K, Rohde C, Young JH, LaVeist TA, Dubay L: Disparities in diabetes: the nexus of race, poverty, and place. Am J Public Health 104:2147–2155, 2014 [PMC free article: PMC4021012] [PubMed: 24228660]
32.
Hamad R, Penko J, Kazi DS, Coxson P, Guzman D, Wei PC, Mason A, Wang EA, Goldman L, Fiscella K, Bibbins-Domingo K: Association of low socioeconomic status with premature coronary heart disease in US adults. JAMA Cardiol 5:899–908, 2020 [PMC free article: PMC7254448] [PubMed: 32459344]
33.
Chamnan P, Simmons RK, Sharp SJ, Griffin SJ, Wareham NJ: Cardiovascular risk assessment scores for people with diabetes: a systematic review. Diabetologia 52:2001–2014, 2009 [PMC free article: PMC2744770] [PubMed: 19629430]
34.
American College of Cardiology. ASCVD Risk Estimator Plus [article online]. Available from https://tools​.acc.org​/ascvd-risk-estimator-plus​/#!/calculate/estimate/. Accessed 3 Mar 2022
35.
Haffner SM, Alexander CM, Cook TJ, Boccuzzi SJ, Musliner TA, Pedersen TR, Kjekshus J, Pyorala K: Reduced coronary events in simvastatin-treated patients with coronary heart disease and diabetes or impaired fasting glucose levels: subgroup analyses in the Scandinavian Simvastatin Survival Study. Arch Intern Med 159:2661–2667, 1999 [PubMed: 10597756]
36.
Stamler J, Vaccaro O, Neaton JD, Wentworth D: Diabetes, other risk factors, and 12-yr cardiovascular mortality for men screened in the Multiple Risk Factor Intervention Trial. Diabetes Care 16:434–444, 1993 [PubMed: 8432214]
37.
Barr EL, Zimmet PZ, Welborn TA, Jolley D, Magliano DJ, Dunstan DW, Cameron AJ, Dwyer T, Taylor HR, Tonkin AM, Wong TY, McNeil J, Shaw JE: Risk of cardiovascular and all-cause mortality in individuals with diabetes mellitus, impaired fasting glucose, and impaired glucose tolerance: the Australian Diabetes, Obesity, and Lifestyle Study (AusDiab). Circulation 116:151–157, 2007 [PubMed: 17576864]
38.
DECODE Study Group; European Diabetes Epidemiology Group: Is the current definition for diabetes relevant to mortality risk from all causes and cardiovascular and noncardiovascular diseases? Diabetes Care 26:688–696, 2003 [PubMed: 12610023]
39.
American Diabetes Association: Standards of medical care in diabetes—2010. Diabetes Care 33(Suppl 1):S11–S61, 2010 [PMC free article: PMC2797382] [PubMed: 20042772]
40.
Khan SS, Butler J, Gheorghiade M: Management of comorbid diabetes mellitus and worsening heart failure. JAMA 311:2379–2380, 2014 [PubMed: 24938559]
41.
Matsushita K, Blecker S, Pazin-Filho A, Bertoni A, Chang PP, Coresh J, Selvin E: The association of hemoglobin A1c with incident heart failure among people without diabetes: the Atherosclerosis Risk in Communities study. Diabetes 59:2020–2026, 2010 [PMC free article: PMC2911067] [PubMed: 20484138]
42.
Selvin E, Steffes MW, Zhu H, Matsushita K, Wagenknecht L, Pankow J, Coresh J, Brancati FL: Glycated hemoglobin, diabetes, and cardiovascular risk in nondiabetic adults. N Engl J Med 362:800–811, 2010 [PMC free article: PMC2872990] [PubMed: 20200384]
43.
Eastwood SV, Tillin T, Sattar N, Forouhi NG, Hughes AD, Chaturvedi N: Associations between prediabetes, by three different diagnostic criteria, and incident CVD differ in South Asians and Europeans. Diabetes Care 38:2325–2332, 2015 [PMC free article: PMC4868252] [PubMed: 26486189]
44.
Barr EL, Boyko EJ, Zimmet PZ, Wolfe R, Tonkin AM, Shaw JE: Continuous relationships between non-diabetic hyperglycaemia and both cardiovascular disease and all-cause mortality: the Australian Diabetes, Obesity, and Lifestyle (AusDiab) study. Diabetologia 52:415–424, 2009 [PubMed: 19130039]
45.
Garg N, Moorthy N, Kapoor A, Tewari S, Kumar S, Sinha A, Shrivastava A, Goel PK: Hemoglobin A(1c) in nondiabetic patients: an independent predictor of coronary artery disease and its severity. Mayo Clin Proc 89:908–916, 2014 [PubMed: 24996234]
46.
Santos-Oliveira R, Purdy C, da Silva MP, dos Anjos Carneiro-Leao AM, Machado M, Einarson TR: Haemoglobin A1c levels and subsequent cardiovascular disease in persons without diabetes: a meta-analysis of prospective cohorts. Diabetologia 54:1327–1334, 2011 [PubMed: 21340623]
47.
Sarwar N, Aspelund T, Eiriksdottir G, Gobin R, Seshasai SR, Forouhi NG, Sigurdsson G, Danesh J, Gudnason V: Markers of dysglycaemia and risk of coronary heart disease in people without diabetes: Reykjavik prospective study and systematic review. PLoS Med 7:e1000278, 2010 [PMC free article: PMC2876150] [PubMed: 20520805]
48.
Parrinello CM, Sharrett AR, Maruthur NM, Bergenstal RM, Grams ME, Coresh J, Selvin E: Racial differences in and prognostic value of biomarkers of hyperglycemia. Diabetes Care 39:589–595, 2016 [PMC free article: PMC4806772] [PubMed: 26681712]
49.
Zaccardi F, Kurl S, Pitocco D, Ronkainen K, Laukkanen JA: Serum fructosamine and risk of cardiovascular and all-cause mortality: a 24-year prospective population-based study. Nutr Metab Cardiovasc Dis 25:236–241, 2015 [PubMed: 25445881]
50.
Ducimetiere P, Eschwege E, Papoz L, Richard JL, Claude JR, Rosselin G: Relationship of plasma insulin levels to the incidence of myocardial infarction and coronary heart disease mortality in a middle-aged population. Diabetologia 19:205–210, 1980 [PubMed: 6997122]
51.
Pyorala K: Relationship of glucose tolerance and plasma insulin to the incidence of coronary heart disease: results from two population studies in Finland. Diabetes Care 2:131–141, 1979 [PubMed: 520116]
52.
Stout RW: Insulin and atheroma. 20-yr perspective. Diabetes Care 13:631–654, 1990 [PubMed: 2192848]
53.
Welborn TA, Wearne K: Coronary heart disease incidence and cardiovascular mortality in Busselton with reference to glucose and insulin concentrations. Diabetes Care 2:154–160, 1979 [PubMed: 520119]
54.
Adler AI, Levy JC, Matthews DR, Stratton IM, Hines G, Holman RR: Insulin sensitivity at diagnosis of type 2 diabetes is not associated with subsequent cardiovascular disease (UKPDS 67). Diabet Med 22:306–311, 2005 [PubMed: 15717879]
55.
Lehto S, Ronnemaa T, Pyorala K, Laakso M: Cardiovascular risk factors clustering with endogenous hyperinsulinaemia predict death from coronary heart disease in patients with type II diabetes. Diabetologia 43:148–155, 2000 [PubMed: 10753035]
56.
Rewers M, Zaccaro D, D’Agostino R, Haffner S, Saad MF, Selby JV, Bergman R, Savage P; Insulin Resistance Atherosclerosis Study Investigators: Insulin sensitivity, insulinemia, and coronary artery disease: the Insulin Resistance Atherosclerosis Study. Diabetes Care 27:781–787, 2004 [PubMed: 14988302]
57.
Ruige JB, Assendelft WJ, Dekker JM, Kostense PJ, Heine RJ, Bouter LM: Insulin and risk of cardiovascular disease: a meta-analysis. Circulation 97:996–1001, 1998 [PubMed: 9529268]
58.
Oh JY, Barrett-Connor E, Wedick NM: Sex differences in the association between proinsulin and intact insulin with coronary heart disease in nondiabetic older adults: the Rancho Bernardo Study. Circulation 105:1311–1316, 2002 [PubMed: 11901041]
59.
Sarwar N, Sattar N, Gudnason V, Danesh J: Circulating concentrations of insulin markers and coronary heart disease: a quantitative review of 19 Western prospective studies. Eur Heart J 28:2491–2497, 2007 [PubMed: 17513304]
60.
Zethelius B, Byberg L, Hales CN, Lithell H, Berne C: Proinsulin is an independent predictor of coronary heart disease: report from a 27-year follow-up study. Circulation 105:2153–2158, 2002 [PubMed: 11994248]
61.
Krauss RM: Lipids and lipoproteins in patients with type 2 diabetes. Diabetes Care 27:1496–1504, 2004 [PubMed: 15161808]
62.
Haffner SM, Stern MP, Hazuda HP, Mitchell BD, Patterson JK: Cardiovascular risk factors in confirmed prediabetic individuals. Does the clock for coronary heart disease start ticking before the onset of clinical diabetes? JAMA 263:2893–2898, 1990 [PubMed: 2338751]
63.
Mora S, Otvos JD, Rosenson RS, Pradhan A, Buring JE, Ridker PM: Lipoprotein particle size and concentration by nuclear magnetic resonance and incident type 2 diabetes in women. Diabetes 59:1153–1160, 2010 [PMC free article: PMC2857895] [PubMed: 20185808]
64.
Festa A, Williams K, Hanley AJ, Otvos JD, Goff DC, Wagenknecht LE, Haffner SM: Nuclear magnetic resonance lipoprotein abnormalities in prediabetic subjects in the Insulin Resistance Atherosclerosis Study. Circulation 111:3465–3472, 2005 [PubMed: 15983261]
65.
Mackey RH, Mora S, Bertoni AG, Wassel CL, Carnethon MR, Sibley CT, Goff DC, Jr.: Lipoprotein particles and incident type 2 diabetes in the Multi-Ethnic Study of Atherosclerosis. Diabetes Care 38:628–636, 2015 [PMC free article: PMC4370328] [PubMed: 25592196]
66.
Laakso M, Voutilainen E, Sarlund H, Aro A, Pyorala K, Penttila I: Serum lipids and lipoproteins in middle-aged non-insulin-dependent diabetics. Atherosclerosis 56:271–281, 1985 [PubMed: 3902037]
67.
Sarwar N, Danesh J, Eiriksdottir G, Sigurdsson G, Wareham N, Bingham S, Boekholdt SM, Khaw KT, Gudnason V: Triglycerides and the risk of coronary heart disease: 10,158 incident cases among 262,525 participants in 29 Western prospective studies. Circulation 115:450–458, 2007 [PubMed: 17190864]
68.
Triglyceride Coronary Disease Genetics Consortium; Emerging Risk Factors Collaboration; Sarwar N, Sandhu MS, Ricketts SL, Butterworth AS, Di Angelantonio E, Boekholdt SM, Ouwehand W, Watkins H, Samani NJ, Saleheen D, Lawlor D, Reilly MP, Hingorani AD, Talmud PJ, Danesh J: Triglyceride-mediated pathways and coronary disease: collaborative analysis of 101 studies. Lancet 375:1634–1639, 2010 [PMC free article: PMC2867029] [PubMed: 20452521]
69.
Rosenson RS, Davidson MH, Hirsh BJ, Kathiresan S, Gaudet D: Genetics and causality of triglyceride-rich lipoproteins in atherosclerotic cardiovascular disease. J Am Coll Cardiol 64:2525–2540, 2014 [PubMed: 25500239]
70.
McPhillips JB, Barrett-Connor E, Wingard DL: Cardiovascular disease risk factors prior to the diagnosis of impaired glucose tolerance and non-insulin-dependent diabetes mellitus in a community of older adults. Am J Epidemiol 131:443–453, 1990 [PubMed: 2301354]
71.
Levin G, Kestenbaum B, Chen YDI, Jacobs DR, Jr., Psaty BM, Rotter JI, Siscovick DS, de Boer IH: Glucose, insulin, and incident hypertension in the Multi-Ethnic Study of Atherosclerosis. Am J Epidemiol 172:1144–1154, 2010 [PMC free article: PMC3004765] [PubMed: 20961972]
72.
Adler AI, Stratton IM, Neil HA, Yudkin JS, Matthews DR, Cull CA, Wright AD, Turner RC, Holman RR: Association of systolic blood pressure with macrovascular and microvascular complications of type 2 diabetes (UKPDS 36): prospective observational study. BMJ 321:412–419, 2000 [PMC free article: PMC27455] [PubMed: 10938049]
73.
Joseph JJ, Deedwania P, Acharya T, Aguilar D, Bhatt DL, Chyun DA, Di Palo KE, Golden SH, Sperling LS; American Heart Association Diabetes Committee of the Council on Lifestyle and Cardiometabolic Health; Council on Arteriosclerosis, Thrombosis and Vascular Biology; Council on Clinical Cardiology; Council on Hypertension: Comprehensive management of cardiovascular risk factors for adults with type 2 diabetes: a Scientific Statement from the American Heart Association. Circulation 145:e722–e759, 2022 [PubMed: 35000404]
74.
Whelton PK, Carey RM, Aronow WS, Casey DE, Jr., Collins KJ, Dennison Himmelfarb C, DePalma SM, Gidding S, Jamerson KA, Jones DW, MacLaughlin EJ, Muntner P, Ovbiagele B, Smith SC, Jr., Spencer CC, Stafford RS, Taler SJ, Thomas RJ, Williams KA, Sr., Williamson JD, Wright JT, Jr.: 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: executive summary: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 138:e426–e483, 2018 [PubMed: 30354655]
75.
ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Das SR, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Kosiborod M, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA: 10. Cardiovascular disease and risk management: standards of care in diabetes—2023. Diabetes Care 46:S158–S190, 2023 [PMC free article: PMC9810475] [PubMed: 36507632]
76.
Barrett-Connor E: The Rancho Bernardo Study: 40 years studying why women have less heart disease than men and how diabetes modifies women’s usual cardiac protection. Glob Heart. 2013 Jun 1 [Epub] doi: 10.1016/j.gheart.2012.12.002 [PMC free article: PMC3810980] [PubMed: 24187655] [CrossRef]
77.
Yusuf S, Hawken S, Ounpuu S, Bautista L, Franzosi MG, Commerford P, Lang CC, Rumboldt Z, Onen CL, Lisheng L, Tanomsup S, Wangai P, Jr., Razak F, Sharma AM, Anand SS; INTERHEART Study Investigators: Obesity and the risk of myocardial infarction in 27,000 participants from 52 countries: a case-control study. Lancet 366:1640–1649, 2005 [PubMed: 16271645]
78.
Araneta MR, Kanaya AM, Hsu WC, Chang HK, Grandinetti A, Boyko EJ, Hayashi T, Kahn SE, Leonetti DL, McNeely MJ, Onishi Y, Sato KK, Fujimoto WY: Optimum BMI cut points to screen Asian Americans for type 2 diabetes. Diabetes Care 38:814–820, 2015 [PMC free article: PMC4407753] [PubMed: 25665815]
79.
Zhang C, Rexrode KM, van Dam RM, Li TY, Hu FB: Abdominal obesity and the risk of all-cause, cardiovascular, and cancer mortality: sixteen years of follow-up in US women. Circulation 117:1658–1667, 2008 [PubMed: 18362231]
80.
de Koning L, Merchant AT, Pogue J, Anand SS: Waist circumference and waist-to-hip ratio as predictors of cardiovascular events: meta-regression analysis of prospective studies. Eur Heart J 28:850–856, 2007 [PubMed: 17403720]
81.
Emerging Risk Factors Collaboration; Wormser D, Kaptoge S, Di Angelantonio E, Wood AM, Pennells L, Thompson A, Sarwar N, Kizer JR, Lawlor DA, Nordestgaard BG, Ridker P, Salomaa V, Stevens J, Woodward M, Sattar N, Collins R, Thompson SG, Whitlock G, Danesh J: Separate and combined associations of body-mass index and abdominal adiposity with cardiovascular disease: collaborative analysis of 58 prospective studies. Lancet 377:1085–1095, 2011 [PMC free article: PMC3145074] [PubMed: 21397319]
82.
Reaven GM: Banting lecture 1988. Role of insulin resistance in human disease. Diabetes 37:1595–1607, 1988 [PubMed: 3056758]
83.
Grundy SM, Cleeman JI, Daniels SR, Donato KA, Eckel RH, Franklin BA, Gordon DJ, Krauss RM, Savage PJ, Smith SC, Jr., Spertus JA, Costa F; American Heart Association; National Heart, Lung, and Blood Institute: Diagnosis and management of the metabolic syndrome: an American Heart Association/National Heart, Lung, and Blood Institute Scientific Statement. Circulation 112:2735–2752, 2005 [PubMed: 16157765]
84.
Alberti KG, Zimmet P, Shaw J; IDF Epidemiology Task Force Consensus Group: The metabolic syndrome—a new worldwide definition. Lancet 366:1059–1062, 2005 [PubMed: 16182882]
85.
Cheung BM, Ong KL, Man YB, Wong LY, Lau CP, Lam KS: Prevalence of the metabolic syndrome in the United States National Health and Nutrition Examination Survey 1999–2002 according to different defining criteria. J Clin Hypertens (Greenwich) 8:562–570, 2006 [PMC free article: PMC8109527] [PubMed: 16896272]
86.
Dekker JM, Girman C, Rhodes T, Nijpels G, Stehouwer CD, Bouter LM, Heine RJ: Metabolic syndrome and 10-year cardiovascular disease risk in the Hoorn Study. Circulation 112:666–673, 2005 [PubMed: 16061755]
87.
Hanley AJ, Karter AJ, Williams K, Festa A, D’Agostino RB, Jr., Wagenknecht LE, Haffner SM: Prediction of type 2 diabetes mellitus with alternative definitions of the metabolic syndrome: the Insulin Resistance Atherosclerosis Study. Circulation 112:3713–3721, 2005 [PubMed: 16344402]
88.
Grundy SM: Does the metabolic syndrome exist? Diabetes Care 29:1689–1692, 2006 [PubMed: 16801603]
89.
Kahn R, Buse J, Ferrannini E, Stern M: The metabolic syndrome: time for a critical appraisal. Joint statement from the American Diabetes Association and the European Association for the Study of Diabetes. Diabetologia 48:1684–1699, 2005 [PubMed: 16079964]
90.
Stern MP, Williams K, Gonzalez-Villalpando C, Hunt KJ, Haffner SM: Does the metabolic syndrome improve identification of individuals at risk of type 2 diabetes and/or cardiovascular disease? Diabetes Care 27:2676–2681, 2004 [PubMed: 15505004]
91.
Diabetes Prevention Program Research Group: Long-term effects of lifestyle intervention or metformin on diabetes development and microvascular complications over 15-year follow-up: the Diabetes Prevention Program Outcomes Study. Lancet Diabetes Endocrinol 3:866–875, 2015 [PMC free article: PMC4623946] [PubMed: 26377054]
92.
Gong Q, Zhang P, Wang J, Ma J, An Y, Chen Y, Zhang B, Feng X, Li H, Chen X, Cheng YJ, Gregg EW, Hu Y, Bennett PH, Li G; Da Qing Diabetes Prevention Study Group: Morbidity and mortality after lifestyle intervention for people with impaired glucose tolerance: 30-year results of the Da Qing Diabetes Prevention Outcome Study. Lancet Diabetes Endocrinol 7:452–461, 2019 [PMC free article: PMC8172050] [PubMed: 31036503]
93.
Look AHEAD Research Group; Gregg EW, Jakicic JM, Blackburn G, Bloomquist P, Bray GA, Clark JM, Coday M, Curtis JM, Egan C, Evans M, Foreyt J, Foster G, Hazuda HP, Hill JO, Horton ES, Hubbard VS, Jeffery RW, Johnson KC, Kitabchi AE, Knowler WC, Kriska A, Lang W, Lewis CE, Montez MG, Nathan DM, Neiberg RH, Patricio J, Peters A, Pi-Sunyer X, Pownall H, Redmon B, Regensteiner J, Rejeski J, Ribisl PM, Safford M, Stewart K, Trence D, Wadden TA, Wing RR, Yanovski SZ: Association of the magnitude of weight loss and changes in physical fitness with long-term cardiovascular disease outcomes in overweight or obese people with type 2 diabetes: a post-hoc analysis of the Look AHEAD randomised clinical trial. Lancet Diabetes Endocrinol 4:913–921, 2016 [PMC free article: PMC5094846] [PubMed: 27595918]
94.
Goldberg RB, Orchard TJ, Crandall JP, Boyko EJ, Budoff M, Dabelea D, Gadde KM, Knowler WC, Lee CG, Nathan DM, Watson K, Temprosa M; Diabetes Prevention Program Research Group: Effects of long-term metformin and lifestyle interventions on cardiovascular events in the Diabetes Prevention Program and its outcome study. Circulation 145:1632–1641, 2022 [PMC free article: PMC9179081] [PubMed: 35603600]
95.
Pradhan AD, Cook NR, Buring JE, Manson JE, Ridker PM: C-reactive protein is independently associated with fasting insulin in nondiabetic women. Arterioscler Thromb Vasc Biol 23:650–655, 2003 [PubMed: 12692007]
96.
Thorand B, Lowel H, Schneider A, Kolb H, Meisinger C, Frohlich M, Koenig W: C-reactive protein as a predictor for incident diabetes mellitus among middle-aged men: results from the MONICA Augsburg cohort study, 1984–1998. Arch Intern Med 163:93–99, 2003 [PubMed: 12523922]
97.
Effoe VS, Correa A, Chen H, Lacy ME, Bertoni AG: High-sensitivity C-reactive protein is associated with incident type 2 diabetes among African Americans: the Jackson Heart Study. Diabetes Care 38:1694–1700, 2015 [PMC free article: PMC4542275] [PubMed: 26068864]
98.
Kanmani S, Kwon M, Shin MK, Kim MK: Association of C-reactive protein with risk of developing type 2 diabetes mellitus, and role of obesity and hypertension: a large population-based Korean cohort study. Sci Rep 9:4573, 2019 [PMC free article: PMC6418102] [PubMed: 30872696]
99.
Ridker PM, Hennekens CH, Buring JE, Rifai N: C-reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women. N Engl J Med 342:836–843, 2000 [PubMed: 10733371]
100.
Quispe R, Michos ED, Martin SS, Puri R, Toth PP, Al Suwaidi J, Banach M, Virani SS, Blumenthal RS, Jones SR, Elshazly MB: High-sensitivity C-reactive protein discordance with atherogenic lipid measures and incidence of atherosclerotic cardiovascular disease in primary prevention: the ARIC study. J Am Heart Assoc 9:e013600, 2020 [PMC free article: PMC7033866] [PubMed: 32013698]
101.
Ridker PM, Buring JE, Rifai N, Cook NR: Development and validation of improved algorithms for the assessment of global cardiovascular risk in women: the Reynolds Risk Score. JAMA 297:611–619, 2007 [PubMed: 17299196]
102.
Ridker PM, Paynter NP, Rifai N, Gaziano JM, Cook NR: C-reactive protein and parental history improve global cardiovascular risk prediction: the Reynolds Risk Score for men. Circulation 118:2243–2251, 2008 [PMC free article: PMC2752381] [PubMed: 18997194]
103.
Ridker PM, Danielson E, Fonseca FA, Genest J, Gotto AM, Jr., Kastelein JJ, Koenig W, Libby P, Lorenzatti AJ, Macfadyen JG, Nordestgaard BG, Shepherd J, Willerson JT, Glynn RJ; JUPITER Trial Study Group: Reduction in C-reactive protein and LDL cholesterol and cardiovascular event rates after initiation of rosuvastatin: a prospective study of the JUPITER trial. Lancet 373:1175–1182, 2009 [PubMed: 19329177]
104.
Gaze DC, Collinson PO: Multiple molecular forms of circulating cardiac troponin: analytical and clinical significance. Ann Clin Biochem 45:349–355, 2008 [PubMed: 18583618]
105.
Park KC, Gaze DC, Collinson PO, Marber MS: Cardiac troponins: from myocardial infarction to chronic disease. Cardiovasc Res 113:1708–1718, 2017 [PMC free article: PMC5852618] [PubMed: 29016754]
106.
Apple FS, Collinson PO; IFCC Task Force on Clinical Applications of Cardiac Biomarkers: Analytical characteristics of high-sensitivity cardiac troponin assays. Clin Chem 58:54–61, 2012 [PubMed: 21965555]
107.
Everett BM, Brooks MM, Vlachos HE, Chaitman BR, Frye RL, Bhatt DL; BARI 2D Study Group: Troponin and cardiac events in stable ischemic heart disease and diabetes. N Engl J Med 373:610–620, 2015 [PMC free article: PMC4627639] [PubMed: 26267622]
108.
Jia X, Sun W, Hoogeveen RC, Nambi V, Matsushita K, Folsom AR, Heiss G, Couper DJ, Solomon SD, Boerwinkle E, Shah A, Selvin E, de Lemos JA, Ballantyne CM: High-sensitivity troponin I and incident coronary events, stroke, heart failure hospitalization, and mortality in the ARIC study. Circulation 139:2642–2653, 2019 [PMC free article: PMC6546524] [PubMed: 31030544]
109.
Ford I, Shah AS, Zhang R, McAllister DA, Strachan FE, Caslake M, Newby DE, Packard CJ, Mills NL: High-sensitivity cardiac troponin, statin therapy, and risk of coronary heart disease. J Am Coll Cardiol 68:2719–2728, 2016 [PMC free article: PMC5176330] [PubMed: 28007133]
110.
Lan NSR, Bell DA, McCaul KA, Vasikaran SD, Yeap BB, Norman PE, Almeida OP, Golledge J, Hankey GJ, Flicker L: High-sensitivity cardiac troponin I improves cardiovascular risk prediction in older men: HIMS (The Health in Men Study). J Am Heart Assoc 8:e011818, 2019 [PMC free article: PMC6474925] [PubMed: 30819029]
111.
Gori M, Gupta DK, Claggett B, Selvin E, Folsom AR, Matsushita K, Bello NA, Cheng S, Shah A, Skali H, Vardeny O, Ni H, Ballantyne CM, Astor BC, Klein BE, Aguilar D, Solomon SD: Natriuretic peptide and high-sensitivity troponin for cardiovascular risk prediction in diabetes: the Atherosclerosis Risk in Communities (ARIC) study. Diabetes Care 39:677–685, 2016 [PMC free article: PMC4839173] [PubMed: 26740635]
112.
Tang O, Matsushita K, Coresh J, Ndumele C, McEvoy JW, Sharrett AR, Hoogeveen R, Ballantyne CM, Selvin E: High-sensitivity cardiac troponin I and T for cardiovascular risk stratification in adults with diabetes. Diabetes Care 43:e144–e146, 2020 [PMC free article: PMC7510022] [PubMed: 32788284]
113.
Daniels LB, Maisel AS: Natriuretic peptides. J Am Coll Cardiol 50:2357–2368, 2007 [PubMed: 18154959]
114.
Goetze JP, Mogelvang R, Maage L, Scharling H, Schnohr P, Sogaard P, Rehfeld JF, Jensen JS: Plasma pro-B-type natriuretic peptide in the general population: screening for left ventricular hypertrophy and systolic dysfunction. Eur Heart J 27:3004–3010, 2006 [PubMed: 17118955]
115.
Nadir MA, Rekhraj S, Wei L, Lim TK, Davidson J, MacDonald TM, Lang CC, Dow E, Struthers AD: Improving the primary prevention of cardiovascular events by using biomarkers to identify individuals with silent heart disease. J Am Coll Cardiol 60:960–968, 2012 [PubMed: 22921971]
116.
Rana BS, Davies JI, Band MM, Pringle SD, Morris A, Struthers AD: B-type natriuretic peptide can detect silent myocardial ischaemia in asymptomatic type 2 diabetes. Heart 92:916–920, 2006 [PMC free article: PMC1860694] [PubMed: 16339811]
117.
Vasan RS, Benjamin EJ, Larson MG, Leip EP, Wang TJ, Wilson PW, Levy D: Plasma natriuretic peptides for community screening for left ventricular hypertrophy and systolic dysfunction: the Framingham Heart Study. JAMA 288:1252–1259, 2002 [PubMed: 12215132]
118.
Wang TJ, Larson MG, Levy D, Benjamin EJ, Leip EP, Omland T, Wolf PA, Vasan RS: Plasma natriuretic peptide levels and the risk of cardiovascular events and death. N Engl J Med 350:655–663, 2004 [PubMed: 14960742]
119.
Nambi V, Liu X, Chambless LE, de Lemos JA, Virani SS, Agarwal S, Boerwinkle E, Hoogeveen RC, Aguilar D, Astor BC, Srinivas PR, Deswal A, Mosley TH, Coresh J, Folsom AR, Heiss G, Ballantyne CM: Troponin T and N-terminal pro-B-type natriuretic peptide: a biomarker approach to predict heart failure risk—the Atherosclerosis Risk in Communities study. Clin Chem 59:1802–1810, 2013 [PMC free article: PMC4208068] [PubMed: 24036936]
120.
Linssen GC, Bakker SJ, Voors AA, Gansevoort RT, Hillege HL, de Jong PE, van Veldhuisen DJ, Gans RO, de Zeeuw D: N-terminal pro-B-type natriuretic peptide is an independent predictor of cardiovascular morbidity and mortality in the general population. Eur Heart J 31:120–127, 2010 [PubMed: 19854731]
121.
Marz W, Tiran B, Seelhorst U, Wellnitz B, Bauersachs J, Winkelmann BR, Boehm BO, Team LS: N-terminal pro-B-type natriuretic peptide predicts total and cardiovascular mortality in individuals with or without stable coronary artery disease: the Ludwigshafen Risk and Cardiovascular Health Study. Clin Chem 53:1075–1083, 2007 [PubMed: 17446333]
122.
Bhalla MA, Chiang A, Epshteyn VA, Kazanegra R, Bhalla V, Clopton P, Krishnaswamy P, Morrison LK, Chiu A, Gardetto N, Mudaliar S, Edelman SV, Henry RR, Maisel AS: Prognostic role of B-type natriuretic peptide levels in patients with type 2 diabetes mellitus. J Am Coll Cardiol 44:1047–1052, 2004 [PubMed: 15337217]
123.
Tsuruda T, Kato J, Sumi T, Mishima K, Masuyama H, Nakao H, Imamura T, Eto T, Kitamura K: Combined use of brain natriuretic peptide and C-reactive protein for predicting cardiovascular risk in outpatients with type 2 diabetes mellitus. Vasc Health Risk Manag 3:417–423, 2007 [PMC free article: PMC2291340] [PubMed: 17969372]
124.
Birukov A, Eichelmann F, Kuxhaus O, Polemiti E, Fritsche A, Wirth J, Boeing H, Weikert C, Schulze MB: Opposing associations of NT-proBNP with risks of diabetes and diabetes-related complications. Diabetes Care 43:2930–2937, 2020 [PMC free article: PMC7770272] [PubMed: 32816995]
125.
Everett BM, Cook NR, Chasman DI, Magnone MC, Bobadilla M, Rifai N, Ridker PM, Pradhan AD: Prospective evaluation of B-type natriuretic peptide concentrations and the risk of type 2 diabetes in women. Clin Chem 59:557–565, 2013 [PMC free article: PMC3694412] [PubMed: 23288489]
126.
Lazo M, Young JH, Brancati FL, Coresh J, Whelton S, Ndumele CE, Hoogeveen R, Ballantyne CM, Selvin E: NH2-terminal pro-brain natriuretic peptide and risk of diabetes. Diabetes 62:3189–3193, 2013 [PMC free article: PMC3749338] [PubMed: 23733199]
127.
Pfister R, Sharp S, Luben R, Welsh P, Barroso I, Salomaa V, Meirhaeghe A, Khaw KT, Sattar N, Langenberg C, Wareham NJ: Mendelian randomization study of B-type natriuretic peptide and type 2 diabetes: evidence of causal association from population studies. PLoS Med 8:e1001112, 2011 [PMC free article: PMC3201934] [PubMed: 22039354]
128.
Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM, Deswal A, Drazner MH, Dunlay SM, Evers LR, Fang JC, Fedson SE, Fonarow GC, Hayek SS, Hernandez AF, Khazanie P, Kittleson MM, Lee CS, Link MS, Milano CA, Nnacheta LC, Sandhu AT, Stevenson LW, Vardeny O, Vest AR, Yancy CW: 2022 AHA/ACC/HFSA guideline for the management of heart failure: executive summary: a report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. J Am Coll Cardiol 79:1757–1780, 2022 [PubMed: 35379504]
129.
Clerico A, Giannoni A, Vittorini S, Emdin M: The paradox of low BNP levels in obesity. Heart Fail Rev 17:81–96, 2012 [PubMed: 21523383]
130.
Wang TJ, Larson MG, Levy D, Benjamin EJ, Leip EP, Wilson PW, Vasan RS: Impact of obesity on plasma natriuretic peptide levels. Circulation 109:594–600, 2004 [PubMed: 14769680]
131.
D’Souza A M, Neumann UH, Glavas MM, Kieffer TJ: The glucoregulatory actions of leptin. Mol Metab 6:1052–1065, 2017 [PMC free article: PMC5605734] [PubMed: 28951828]
132.
Susairaj P, Snehalatha C, Nanditha A, Satheesh K, Raghavan A, Vinitha R, Ramachandran A: Analysis of an Indian diabetes prevention programme on association of adipokines and a hepatokine with incident diabetes. Sci Rep 11:20327, 2021 [PMC free article: PMC8514464] [PubMed: 34645898]
133.
Wannamethee SG, Lowe GD, Rumley A, Cherry L, Whincup PH, Sattar N: Adipokines and risk of type 2 diabetes in older men. Diabetes Care 30:1200–1205, 2007 [PubMed: 17322479]
134.
Muse ED, Feldman DI, Blaha MJ, Dardari ZA, Blumenthal RS, Budoff MJ, Nasir K, Criqui MH, Cushman M, McClelland RL, Allison MA: The association of resistin with cardiovascular disease in the Multi-Ethnic Study of Atherosclerosis. Atherosclerosis 239:101–108, 2015 [PMC free article: PMC4331252] [PubMed: 25585029]
135.
Pischon T, Girman CJ, Hotamisligil GS, Rifai N, Hu FB, Rimm EB: Plasma adiponectin levels and risk of myocardial infarction in men. JAMA 291:1730–1737, 2004 [PubMed: 15082700]
136.
Landecho MF, Tuero C, Valenti V, Bilbao I, de la Higuera M, Fruhbeck G: Relevance of leptin and other adipokines in obesity-associated cardiovascular risk. Nutrients 11:2664, 2019 [PMC free article: PMC6893824] [PubMed: 31694146]
137.
Martin SS, Blaha MJ, Muse ED, Qasim AN, Reilly MP, Blumenthal RS, Nasir K, Criqui MH, McClelland RL, Hughes-Austin JM, Allison MA: Leptin and incident cardiovascular disease: the Multi-Ethnic Study of Atherosclerosis (MESA). Atherosclerosis 239:67–72, 2015 [PMC free article: PMC4331218] [PubMed: 25574859]
138.
Hansen D, Dendale P, Beelen M, Jonkers RA, Mullens A, Corluy L, Meeusen R, van Loon LJ: Plasma adipokine and inflammatory marker concentrations are altered in obese, as opposed to non-obese, type 2 diabetes patients. Eur J Appl Physiol 109:397–404, 2010 [PMC free article: PMC2874484] [PubMed: 20131064]
139.
Sattar N, Wannamethee G, Sarwar N, Tchernova J, Cherry L, Wallace AM, Danesh J, Whincup PH: Adiponectin and coronary heart disease: a prospective study and meta-analysis. Circulation 114:623–629, 2006 [PubMed: 16894037]
140.
Schmidt MI, Duncan BB, Vigo A, Pankow JS, Couper D, Ballantyne CM, Hoogeveen RC, Heiss G; ARIC Investigators: Leptin and incident type 2 diabetes: risk or protection? Diabetologia 49:2086–2096, 2006 [PubMed: 16850292]
141.
Bidulescu A, Dinh PC, Jr., Sarwary S, Forsyth E, Luetke MC, King DB, Liu J, Davis SK, Correa A: Associations of leptin and adiponectin with incident type 2 diabetes and interactions among African Americans: the Jackson Heart Study. BMC Endocr Disord 20:31, 2020 [PMC free article: PMC7057597] [PubMed: 32131811]
142.
Sattar N, Wannamethee G, Sarwar N, Chernova J, Lawlor DA, Kelly A, Wallace AM, Danesh J, Whincup PH: Leptin and coronary heart disease: prospective study and systematic review. J Am Coll Cardiol 53:167–175, 2009 [PubMed: 19130985]
143.
Yang H, Guo W, Li J, Cao S, Zhang J, Pan J, Wang Z, Wen P, Shi X, Zhang S: Leptin concentration and risk of coronary heart disease and stroke: a systematic review and meta-analysis. PLoS One 12:e0166360, 2017 [PMC free article: PMC5344319] [PubMed: 28278178]
144.
Rawshani A, Rawshani A, Franzen S, Sattar N, Eliasson B, Svensson AM, Zethelius B, Miftaraj M, McGuire DK, Rosengren A, Gudbjornsdottir S: Risk factors, mortality, and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 379:633–644, 2018 [PubMed: 30110583]
145.
Budoff MJ, Raggi P, Beller GA, Berman DS, Druz RS, Malik S, Rigolin VH, Weigold WG, Soman P; Imaging Council of the American College of Cardiology: Noninvasive cardiovascular risk assessment of the asymptomatic diabetic patient: the Imaging Council of the American College of Cardiology. JACC Cardiovasc Imaging 9:176–192, 2016 [PMC free article: PMC5371352] [PubMed: 26846937]
146.
Michos ED, Blaha MJ, Blumenthal RS: Use of the coronary artery calcium score in discussion of initiation of statin therapy in primary prevention. Mayo Clin Proc 92:1831–1841, 2017 [PubMed: 29108840]
147.
Budoff M, Backlund JC, Bluemke DA, Polak J, Bebu I, Schade D, Strowig S, Raskin P, Lachin JM; DCCT/EDIC Research Group: The association of coronary artery calcification with subsequent incidence of cardiovascular disease in type 1 diabetes: the DCCT/EDIC trials. JACC Cardiovasc Imaging 12:1341–1349, 2019 [PMC free article: PMC6612565] [PubMed: 30878435]
148.
Yeboah J, McClelland RL, Polonsky TS, Burke GL, Sibley CT, O’Leary D, Carr JJ, Goff DC, Greenland P, Herrington DM: Comparison of novel risk markers for improvement in cardiovascular risk assessment in intermediate-risk individuals. JAMA 308:788–795, 2012 [PMC free article: PMC4141475] [PubMed: 22910756]
149.
Yeboah J, Young R, McClelland RL, Delaney JC, Polonsky TS, Dawood FZ, Blaha MJ, Miedema MD, Sibley CT, Carr JJ, Burke GL, Goff DC, Jr., Psaty BM, Greenland P, Herrington DM: Utility of nontraditional risk markers in atherosclerotic cardiovascular disease risk assessment. J Am Coll Cardiol 67:139–147, 2016 [PMC free article: PMC4724058] [PubMed: 26791059]
150.
Blaha MJ, Cainzos-Achirica M, Greenland P, McEvoy JW, Blankstein R, Budoff MJ, Dardari Z, Sibley CT, Burke GL, Kronmal RA, Szklo M, Blumenthal RS, Nasir K: Role of coronary artery calcium score of zero and other negative risk markers for cardiovascular disease: the Multi-Ethnic Study of Atherosclerosis (MESA). Circulation 133:849–858, 2016 [PMC free article: PMC4775391] [PubMed: 26801055]
151.
Malik S, Zhao Y, Budoff M, Nasir K, Blumenthal RS, Bertoni AG, Wong ND: 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 2:1332–1340, 2017 [PMC free article: PMC5814996] [PubMed: 29117273]
152.
Wong ND, Cordola Hsu AR, Rozanski A, Shaw LJ, Whelton SP, Budoff MJ, Nasir K, Miedema MD, Rumberger J, Blaha MJ, Berman DS: Sex differences in coronary artery calcium and mortality from coronary heart disease, cardiovascular disease, and all causes in adults with diabetes: the Coronary Calcium Consortium. Diabetes Care 43:2597–2606, 2020 [PMC free article: PMC8051260] [PubMed: 32816996]
153.
Al Rifai M, Blaha MJ, Nambi V, Shea SJC, Michos ED, Blumenthal RS, Ballantyne CM, Szklo M, Greenland P, Miedema MD, Nasir K, Rotter JI, Guo X, Yao J, Post WS, Virani SS: Determinants of incident atherosclerotic cardiovascular disease events among those with absent coronary artery calcium: Multi-Ethnic Study of Atherosclerosis. Circulation 145:259–267, 2022 [PMC free article: PMC8792296] [PubMed: 34879218]
154.
Valenti V, Hartaigh BO, Cho I, Schulman-Marcus J, Gransar H, Heo R, Truong QA, Shaw LJ, Knapper J, Kelkar AA, Sciarretta S, Chang HJ, Callister TQ, Min JK: Absence of coronary artery calcium identifies asymptomatic diabetic individuals at low near-term but not long-term risk of mortality: a 15-year follow-up study of 9715 patients. Circ Cardiovasc Imaging 9:e003528, 2016 [PMC free article: PMC4745994] [PubMed: 26848062]
155.
Dzaye O, Dardari ZA, Cainzos-Achirica M, Blankstein R, Agatston AS, Duebgen M, Yeboah J, Szklo M, Budoff MJ, Lima JAC, Blumenthal RS, Nasir K, Blaha MJ: Warranty period of a calcium score of zero: comprehensive analysis from MESA. JACC Cardiovasc Imaging 14:990–1002, 2021 [PMC free article: PMC8076346] [PubMed: 33129734]
156.
Wong ND, Nelson JC, Granston T, Bertoni AG, Blumenthal RS, Carr JJ, Guerci A, Jacobs DR, Jr., Kronmal R, Liu K, Saad M, Selvin E, Tracy R, Detrano R: Metabolic syndrome, diabetes, and incidence and progression of coronary calcium: the Multiethnic Study of Atherosclerosis study. JACC Cardiovasc Imaging 5:358–366, 2012 [PMC free article: PMC3327555] [PubMed: 22498324]
157.
Kowall B, Lehmann N, Mahabadi AA, Moebus S, Budde T, Seibel R, Gronemeyer D, Erbel R, Jockel KH, Stang A: Progression of coronary artery calcification is stronger in poorly than in well controlled diabetes: results from the Heinz Nixdorf Recall Study. J Diabetes Complications 31:234–240, 2017 [PubMed: 27665253]
158.
Razavi AC, Wong N, Budoff M, Bazzano LA, Kelly TN, He J, Fernandez C, Lima J, Polak JF, Mongraw-Chaffin M, deFilippi C, Szklo M, Bertoni AG, Blumenthal RS, Blaha MJ, Whelton SP: Predicting long-term absence of coronary artery calcium in metabolic syndrome and diabetes: the MESA study. JACC Cardiovasc Imaging 14:219–229, 2021 [PMC free article: PMC7796947] [PubMed: 33129732]
159.
Diabetes Control and Complications Trial Research Group; Nathan DM, Genuth S, Lachin J, Cleary P, Crofford O, Davis M, Rand L, Siebert C: The effect of intensive treatment of diabetes on the development and progression of long-term complications in insulin-dependent diabetes mellitus. N Engl J Med 329:977–986, 1993 [PubMed: 8366922]
160.
Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group. Lancet 352:837–853, 1998 [PubMed: 9742976]
161.
Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. Lancet 352:854–865, 1998 [PubMed: 9742977]
162.
ADVANCE Collaborative Group; Patel A, MacMahon S, Chalmers J, Neal B, Billot L, Woodward M, Marre M, Cooper M, Glasziou P, Grobbee D, Hamet P, Harrap S, Heller S, Liu L, Mancia G, Mogensen CE, Pan C, Poulter N, Rodgers A, Williams B, Bompoint S, de Galan BE, Joshi R, Travert F: Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N Engl J Med 358:2560–2572, 2008 [PubMed: 18539916]
163.
Action to Control Cardiovascular Risk in Diabetes Study Group; Gerstein HC, Miller ME, Byington RP, Goff DC, Jr., Bigger JT, Buse JB, Cushman WC, Genuth S, Ismail-Beigi F, Grimm RH, Jr., Probstfield JL, Simons-Morton DG, Friedewald WT: Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 358:2545–2559, 2008 [PMC free article: PMC4551392] [PubMed: 18539917]
164.
Duckworth W, Abraira C, Moritz T, Reda D, Emanuele N, Reaven PD, Zieve FJ, Marks J, Davis SN, Hayward R, Warren SR, Goldman S, McCarren M, Vitek ME, Henderson WG, Huang GD; VADT Investigators: Glucose control and vascular complications in veterans with type 2 diabetes. N Engl J Med 360:129–139, 2009 [PubMed: 19092145]
165.
Diabetes Control and Complications Trial (DCCT)/Epidemiology of Diabetes Interventions and Complications (EDIC) Study Research Group: Intensive diabetes treatment and cardiovascular outcomes in type 1 diabetes: the DCCT/EDIC Study 30-year follow-up. Diabetes Care 39:686–693, 2016
166.
Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA: 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med 359:1577–1589, 2008 [PubMed: 18784090]
167.
Reaven PD, Emanuele NV, Wiitala WL, Bahn GD, Reda DJ, McCarren M, Duckworth WC, Hayward RA; VADT Investigators: Intensive glucose control in patients with type 2 diabetes—5-year follow-up. N Engl J Med 380:2215–2224, 2019 [PMC free article: PMC6706253] [PubMed: 31167051]
168.
Boussageon R, Bejan-Angoulvant T, Saadatian-Elahi M, Lafont S, Bergeonneau C, Kassai B, Erpeldinger S, Wright JM, Gueyffier F, Cornu C: Effect of intensive glucose lowering treatment on all cause mortality, cardiovascular death, and microvascular events in type 2 diabetes: meta-analysis of randomised controlled trials. BMJ 343:d4169, 2011 [PMC free article: PMC3144314] [PubMed: 21791495]
169.
Malmberg K, Ryden L, Efendic S, Herlitz J, Nicol P, Waldenstrom A, Wedel H, Welin L: Randomized trial of insulin-glucose infusion followed by subcutaneous insulin treatment in diabetic patients with acute myocardial infarction (DIGAMI study): effects on mortality at 1 year. J Am Coll Cardiol 26:57–65, 1995 [PubMed: 7797776]
170.
Malmberg K, Ryden L, Wedel H, Birkeland K, Bootsma A, Dickstein K, Efendic S, Fisher M, Hamsten A, Herlitz J, Hildebrandt P, MacLeod K, Laakso M, Torp-Pedersen C, Waldenstrom A; DIGAMI 2 Investigators: Intense metabolic control by means of insulin in patients with diabetes mellitus and acute myocardial infarction (DIGAMI 2): effects on mortality and morbidity. Eur Heart J 26:650–661, 2005 [PubMed: 15728645]
171.
van den Berghe G, Wouters P, Weekers F, Verwaest C, Bruyninckx F, Schetz M, Vlasselaers D, Ferdinande P, Lauwers P, Bouillon R: Intensive insulin therapy in critically ill patients. N Engl J Med 345:1359–1367, 2001 [PubMed: 11794168]
172.
van den Berghe G, Wilmer A, Hermans G, Meersseman W, Wouters PJ, Milants I, Van Wijngaerden E, Bobbaers H, Bouillon R: Intensive insulin therapy in the medical ICU. N Engl J Med 354:449–461, 2006 [PubMed: 16452557]
173.
NICE-SUGAR Study Investigators; Finfer S, Chittock DR, Su SY, Blair D, Foster D, Dhingra V, Bellomo R, Cook D, Dodek P, Henderson WR, Hebert PC, Heritier S, Heyland DK, McArthur C, McDonald E, Mitchell I, Myburgh JA, Norton R, Potter J, Robinson BG, Ronco JJ: Intensive versus conventional glucose control in critically ill patients. N Engl J Med 360:1283–1297, 2009 [PubMed: 19318384]
174.
Ference BA, Graham I, Tokgozoglu L, Catapano AL: Impact of lipids on cardiovascular health: JACC Health Promotion Series. J Am Coll Cardiol 72:1141–1156, 2018 [PubMed: 30165986]
175.
Michos ED, McEvoy JW, Blumenthal RS: Lipid management for the prevention of atherosclerotic cardiovascular disease. N Engl J Med 381:1557–1567, 2019 [PubMed: 31618541]
176.
Cholesterol Treatment Trialists’ (CTT) Collaborators; Kearney PM, Blackwell L, Collins R, Keech A, Simes J, Peto R, Armitage J, Baigent C: Efficacy of cholesterol-lowering therapy in 18,686 people with diabetes in 14 randomised trials of statins: a meta-analysis. Lancet 371:117–125, 2008 [PubMed: 18191683]
177.
Sattar N, Preiss D, Murray HM, Welsh P, Buckley BM, de Craen AJ, Seshasai SR, McMurray JJ, Freeman DJ, Jukema JW, Macfarlane PW, Packard CJ, Stott DJ, Westendorp RG, Shepherd J, Davis BR, Pressel SL, Marchioli R, Marfisi RM, Maggioni AP, Tavazzi L, Tognoni G, Kjekshus J, Pedersen TR, Cook TJ, Gotto AM, Clearfield MB, Downs JR, Nakamura H, Ohashi Y, Mizuno K, Ray KK, Ford I: Statins and risk of incident diabetes: a collaborative meta-analysis of randomised statin trials. Lancet 375:735–742, 2010 [PubMed: 20167359]
178.
Cai R, Yuan Y, Sun J, Xia W, Huang R, Tian S, Dong X, Shen Y, Wang S: Statins worsen glycemic control of T2DM in target LDL-c level and LDL-c reduction dependent manners: a meta-analysis. Expert Opin Pharmacother 17:1839–1849, 2016 [PubMed: 27488607]
179.
Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, Braun LT, de Ferranti S, Faiella-Tommasino J, Forman DE, Goldberg R, Heidenreich PA, Hlatky MA, Jones DW, Lloyd-Jones D, Lopez-Pajares N, Ndumele CE, Orringer CE, Peralta CA, Saseen JJ, Smith SC, Jr., Sperling L, Virani SS, Yeboah J: 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. Circulation 139:e1046–e1081, 2019 [PubMed: 30565953]
180.
Giugliano RP, Cannon CP, Blazing MA, Nicolau JC, Corbalan R, Spinar J, Park JG, White JA, Bohula EA, Braunwald E; IMPROVE-IT 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 137:1571–1582, 2018 [PubMed: 29263150]
181.
Sabatine MS, Leiter LA, Wiviott SD, Giugliano RP, Deedwania P, De Ferrari GM, Murphy SA, Kuder JF, Gouni-Berthold I, Lewis BS, Handelsman Y, Pineda AL, Honarpour N, Keech AC, Sever PS, Pedersen TR: 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 5:941–950, 2017 [PubMed: 28927706]
182.
Ray KK, Colhoun HM, Szarek M, Baccara-Dinet M, Bhatt DL, Bittner VA, Budaj AJ, Diaz R, Goodman SG, Hanotin C, Harrington RA, Jukema JW, Loizeau V, Lopes RD, Moryusef A, Murin J, Pordy R, Ristic AD, Roe MT, Tunon J, White HD, Zeiher AM, Schwartz GG, Steg PG; 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 7:618–628, 2019 [PubMed: 31272931]
183.
Wright RS, Ray KK, Raal FJ, Kallend DG, Jaros M, Koenig W, Leiter LA, Landmesser U, Schwartz GG, Friedman A, Wijngaard PLJ, Garcia Conde L, Kastelein JJP; ORION Phase III Investigators: Pooled patient-level analysis of inclisiran trials in patients with familial hypercholesterolemia or atherosclerosis. J Am Coll Cardiol 77:1182–1193, 2021 [PubMed: 33663735]
184.
Leiter LA, Teoh H, Kallend D, Wright RS, Landmesser U, Wijngaard PLJ, Kastelein JJP, Ray KK: Inclisiran lowers LDL-C and PCSK9 irrespective of diabetes status: the ORION-1 randomized clinical trial. Diabetes Care 42:173–176, 2019 [PubMed: 30487231]
185.
Leiter LA, Kallend DG, Koenig W, Landmesser U, Raal FJ, Ray KK, Schwartz GG, Chiang Y, Garcia Conde Orozco L, Wright RS: Efficacy and safety of inclisiran by baseline glycemic status: a post hoc pooled analysis of the ORION-10 and ORION-11 phase III randomized controlled trials (Abstract). Circulation 144(Suppl 1):A11033, 2021. doi:10.1161/circ.144.suppl_1.11033 [CrossRef]
186.
Leiter LA, Banach M, Catapano AL, Duell PB, Gotto AM, Jr., Laufs U, Mancini GBJ, Ray KK, Hanselman JC, Ye Z, Bays HE: Bempedoic acid in patients with type 2 diabetes mellitus, prediabetes, and normoglycaemia: a post hoc analysis of efficacy and glycaemic control using pooled data from phase 3 clinical trials. Diabetes Obes Metab 24:868–880, 2022 [PMC free article: PMC9306638] [PubMed: 34981622]
187.
Masson W, Lobo M, Lavalle-Cobo A, Masson G, Molinero G: Effect of bempedoic acid on new onset or worsening diabetes: a meta-analysis. Diabetes Res Clin Pract 168:108369, 2020 [PubMed: 32827596]
188.
Nissen SE, Lincoff AM, Brennan D, Ray KK, Mason D, Kastelein JJP, Thompson PD, Libby P, Cho L, Plutzky J, Bays HE, Moriarty PM, Menon V, Grobbee DE, Louie MJ, Chen C-F, Li N, Bloedon L, Robinson P, Horner M, Sasiela WJ, McCluskey J, Davey D, Fajardo-Campos P, Petrovic P, Fedacko J, Zmuda W, Lukyanov Y, Nicholls SJ; CLEAR Outcomes Investigators. Bempedoic acid and cardiovascular outcomes in statin-intolerant patients. N Engl J Med 388:1353–1364, 2023 [PubMed: 36876740]
189.
Bhatt DL, Steg PG, Miller M, Brinton EA, Jacobson TA, Ketchum SB, Doyle RT, Jr., Juliano RA, Jiao L, Granowitz C, Tardif JC, Ballantyne CM; REDUCE-IT Investigators: Cardiovascular risk reduction with icosapent ethyl for hypertriglyceridemia. N Engl J Med 380:11–22, 2019 [PubMed: 30415628]
190.
Bhatt DL, Brinton EA, Miller M, Steg PG, Jacobson TA, Ketchum SB, Doyle RT, Juliano RA, Jiao L, Granowitz C, Ganda O, Welty FK, Busch RS, Goldberg AC, Herrington DM, Budoff M, Tardif J, Ballantyne CM: Substantial cardiovascular benefit from icosapent ethyl in patients with diabetes: REDUCE-IT DIABETES (Abstract). 80th Scientific Sessions of the American Diabetes Association, June 12–16, 2020. doi: 10.2337/db20-4-LB10.2337/db20-4-LB [CrossRef] [CrossRef]
191.
Nicholls SJ, Lincoff AM, Garcia M, Bash D, Ballantyne CM, Barter PJ, Davidson MH, Kastelein JJP, Koenig W, McGuire DK, Mozaffarian D, Ridker PM, Ray KK, Katona BG, Himmelmann A, Loss LE, Rensfeldt M, Lundstrom T, Agrawal R, Menon V, Wolski K, Nissen SE: 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 324:2268–2280, 2020 [PMC free article: PMC7667577] [PubMed: 33190147]
192.
Kalstad AA, Myhre PL, Laake K, Tveit SH, Schmidt EB, Smith P, Nilsen DWT, Tveit A, Fagerland MW, Solheim S, Seljeflot I, Arnesen H; OMEMI Investigators: Effects of n-3 fatty acid supplements in elderly patients after myocardial infarction: a randomized, controlled trial. Circulation 143:528–539, 2021 [PubMed: 33191772]
193.
Aung T, Halsey J, Kromhout D, Gerstein HC, Marchioli R, Tavazzi L, Geleijnse JM, Rauch B, Ness A, Galan P, Chew EY, Bosch J, Collins R, Lewington S, Armitage J, Clarke R; Omega-3 Treatment Trialists’ Collaboration: Associations of omega-3 fatty acid supplement use with cardiovascular disease risks: meta-analysis of 10 trials involving 77 917 individuals. JAMA Cardiol 3:225–234, 2018 [PMC free article: PMC5885893] [PubMed: 29387889]
194.
Nicholls SJ, Lincoff AM, Bash D, Ballantyne CM, Barter PJ, Davidson MH, Kastelein JJP, Koenig W, McGuire DK, Mozaffarian D, Pedersen TR, Ridker PM, Ray K, Karlson BW, Lundstrom T, Wolski K, Nissen SE: Assessment of omega-3 carboxylic acids in statin-treated patients with high levels of triglycerides and low levels of high-density lipoprotein cholesterol: rationale and design of the STRENGTH trial. Clin Cardiol 41:1281–1288, 2018 [PMC free article: PMC6489732] [PubMed: 30125052]
195.
Rubins HB, Robins SJ, Collins D, Fye CL, Anderson JW, Elam MB, Faas FH, Linares E, Schaefer EJ, Schectman G, Wilt TJ, Wittes J: Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. N Engl J Med 341:410–418, 1999 [PubMed: 10438259]
196.
Keech A, Simes RJ, Barter P, Best J, Scott R, Taskinen MR, Forder P, Pillai A, Davis T, Glasziou P, Drury P, Kesaniemi YA, Sullivan D, Hunt D, Colman P, d’Emden M, Whiting M, Ehnholm C, Laakso M; 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 366:1849–1861, 2005 [PubMed: 16310551]
197.
ACCORD Study Group; Ginsberg HN, Elam MB, Lovato LC, Crouse JR, 3rd, Leiter LA, Linz P, Friedewald WT, Buse JB, Gerstein HC, Probstfield J, Grimm RH, Ismail-Beigi F, Bigger JT, Goff DC, Jr., Cushman WC, Simons-Morton DG, Byington RP: Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med 362:1563–1574, 2010 [PMC free article: PMC2879499] [PubMed: 20228404]
198.
Das Pradhan A, Glynn RJ, Fruchart JC, MacFadyen JG, Zaharris ES, Everett BM, Campbell SE, Oshima R, Amarenco P, Blom DJ, Brinton EA, Eckel RH, Elam MB, Felicio JS, Ginsberg HN, Goudev A, Ishibashi S, Joseph J, Kodama T, Koenig W, Leiter LA, Lorenzatti AJ, Mankovsky B, Marx N, Nordestgaard BG, Pall D, Ray KK, Santos RD, Soran H, Susekov A, Tendera M, Yokote K, Paynter NP, Buring JE, Libby P, Ridker PM; PROMINENT Investigators: Triglyceride lowering with pemafibrate to reduce cardiovascular risk. N Engl J Med 387:1923–1934, 2022 [PubMed: 36342113]
199.
Hansson L, Zanchetti A, Carruthers SG, Dahlof B, Elmfeldt D, Julius S, Menard J, Rahn KH, Wedel H, Westerling S: Effects of intensive blood-pressure lowering and low-dose aspirin in patients with hypertension: principal results of the Hypertension Optimal Treatment (HOT) randomised trial. HOT Study Group. Lancet 351:1755–1762, 1998 [PubMed: 9635947]
200.
Patel A; ADVANCE Collaborative Group; MacMahon S, Chalmers J, Neal B, Woodward M, Billot L, Harrap S, Poulter N, Marre M, Cooper M, Glasziou P, Grobbee DE, Hamet P, Heller S, Liu LS, Mancia G, Mogensen CE, Pan CY, Rodgers A, Williams B: 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 370:829–840, 2007 [PubMed: 17765963]
201.
de Galan BE: Blood pressure control in type 2 diabetes. N Engl J Med 363:695–696, 2010 [PubMed: 20842774]
202.
ACCORD Study Group; Cushman WC, Evans GW, Byington RP, Goff DC, Jr., Grimm RH, Jr., Cutler JA, Simons-Morton DG, Basile JN, Corson MA, Probstfield JL, Katz L, Peterson KA, Friedewald WT, Buse JB, Bigger JT, Gerstein HC, Ismail-Beigi F: Effects of intensive blood-pressure control in type 2 diabetes mellitus. N Engl J Med 362:1575–1585, 2010 [PMC free article: PMC4123215] [PubMed: 20228401]
203.
SPRINT Research Group; Wright JT, Jr., Williamson JD, Whelton PK, Snyder JK, Sink KM, Rocco MV, Reboussin DM, Rahman M, Oparil S, Lewis CE, Kimmel PL, Johnson KC, Goff DC, Jr., Fine LJ, Cutler JA, Cushman WC, Cheung AK, Ambrosius WT: A randomized trial of intensive versus standard blood-pressure control. N Engl J Med 373:2103–2116, 2015 [PMC free article: PMC4689591] [PubMed: 26551272]
204.
Zhang W, Zhang S, Deng Y, Wu S, Ren J, Sun G, Yang J, Jiang Y, Xu X, Wang TD, Chen Y, Li Y, Yao L, Li D, Wang L, Shen X, Yin X, Liu W, Zhou X, Zhu B, Guo Z, Liu H, Chen X, Feng Y, Tian G, Gao X, Kario K, Cai J; STEP Study Group: Trial of intensive blood-pressure control in older patients with hypertension. N Engl J Med 385:1268–1279, 2021 [PubMed: 34491661]
205.
Capodanno D, Angiolillo DJ: Antithrombotic therapy for atherosclerotic cardiovascular disease risk mitigation in patients with coronary artery disease and diabetes mellitus. Circulation 142:2172–2188, 2020 [PubMed: 33253005]
206.
Antithrombotic Trialists’ Collaboration; Baigent C, Blackwell L, Collins R, Emberson J, Godwin J, Peto R, Buring J, Hennekens C, Kearney P, Meade T, Patrono C, Roncaglioni MC, Zanchetti A: Aspirin in the primary and secondary prevention of vascular disease: collaborative meta-analysis of individual participant data from randomised trials. Lancet 373:1849–1860, 2009 [PMC free article: PMC2715005] [PubMed: 19482214]
207.
Ogawa H, Nakayama M, Morimoto T, Uemura S, Kanauchi M, Doi N, Jinnouchi H, Sugiyama S, Saito Y; Japanese Primary Prevention of Atherosclerosis With Aspirin for Diabetes (JPAD) Trial Investigators: Low-dose aspirin for primary prevention of atherosclerotic events in patients with type 2 diabetes: a randomized controlled trial. JAMA 300:2134–2141, 2008 [PubMed: 18997198]
208.
Saito Y, Okada S, Ogawa H, Soejima H, Sakuma M, Nakayama M, Doi N, Jinnouchi H, Waki M, Masuda I, Morimoto T; JPAD Trial Investigators: Low-dose aspirin for primary prevention of cardiovascular events in patients with type 2 diabetes mellitus: 10-year follow-up of a randomized controlled trial. Circulation 135:659–670, 2017 [PubMed: 27881565]
209.
De Berardis G, Sacco M, Strippoli GF, Pellegrini F, Graziano G, Tognoni G, Nicolucci A: Aspirin for primary prevention of cardiovascular events in people with diabetes: meta-analysis of randomised controlled trials. BMJ 339:b4531, 2009 [PMC free article: PMC2774388] [PubMed: 19897665]
210.
ASCEND Study Collaborative Group; Bowman L, Mafham M, Wallendszus K, Stevens W, Buck G, Barton J, Murphy K, Aung T, Haynes R, Cox J, Murawska A, Young A, Lay M, Chen F, Sammons E, Waters E, Adler A, Bodansky J, Farmer A, McPherson R, Neil A, Simpson D, Peto R, Baigent C, Collins R, Parish S, Armitage J: Effects of aspirin for primary prevention in persons with diabetes mellitus. N Engl J Med 379:1529–1539, 2018 [PubMed: 30146931]
211.
Gaziano JM, Brotons C, Coppolecchia R, Cricelli C, Darius H, Gorelick PB, Howard G, Pearson TA, Rothwell PM, Ruilope LM, Tendera M, Tognoni G; 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 392:1036–1046, 2018 [PMC free article: PMC7255888] [PubMed: 30158069]
212.
McNeil JJ, Wolfe R, Woods RL, Tonkin AM, Donnan GA, Nelson MR, Reid CM, Lockery JE, Kirpach B, Storey E, Shah RC, Williamson JD, Margolis KL, Ernst ME, Abhayaratna WP, Stocks N, Fitzgerald SM, Orchard SG, Trevaks RE, Beilin LJ, Johnston CI, Ryan J, Radziszewska B, Jelinek M, Malik M, Eaton CB, Brauer D, Cloud G, Wood EM, Mahady SE, Satterfield S, Grimm R, Murray AM; ASPREE Investigator Group: Effect of aspirin on cardiovascular events and bleeding in the healthy elderly. N Engl J Med 379:1509–1518, 2018 [PMC free article: PMC6289056] [PubMed: 30221597]
213.
McNeil JJ, Nelson MR, Woods RL, Lockery JE, Wolfe R, Reid CM, Kirpach B, Shah RC, Ives DG, Storey E, Ryan J, Tonkin AM, Newman AB, Williamson JD, Margolis KL, Ernst ME, Abhayaratna WP, Stocks N, Fitzgerald SM, Orchard SG, Trevaks RE, Beilin LJ, Donnan GA, Gibbs P, Johnston CI, Radziszewska B, Grimm R, Murray AM; ASPREE Investigator Group: Effect of aspirin on all-cause mortality in the healthy elderly. N Engl J Med 379:1519–1528, 2018 [PMC free article: PMC6433466] [PubMed: 30221595]
214.
Zheng SL, Roddick AJ: Association of aspirin use for primary prevention with cardiovascular events and bleeding events: a systematic review and meta-analysis. JAMA 321:277–287, 2019 [PMC free article: PMC6439678] [PubMed: 30667501]
215.
Caldeira D, Alves M, David C, Costa J, Ferreira JJ, Pinto FJ: Aspirin in the primary prevention of cardiovascular disease on diabetic patients: systematic review and meta-analysis. Prim Care Diabetes 14:213–221, 2020 [PubMed: 31791903]
216.
Gaede P, Vedel P, Larsen N, Jensen GV, Parving HH, Pedersen O: Multifactorial intervention and cardiovascular disease in patients with type 2 diabetes. N Engl J Med 348:383–393, 2003 [PubMed: 12556541]
217.
Gaede P, Lund-Andersen H, Parving HH, Pedersen O: Effect of a multifactorial intervention on mortality in type 2 diabetes. N Engl J Med 358:580–591, 2008 [PubMed: 18256393]
218.
Umpierre D, Ribeiro PA, Kramer CK, Leitao CB, Zucatti AT, Azevedo MJ, Gross JL, Ribeiro JP, Schaan BD: Physical activity advice only or structured exercise training and association with HbA1c levels in type 2 diabetes: a systematic review and meta-analysis. JAMA 305:1790–1799, 2011 [PubMed: 21540423]
219.
Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, Nathan DM; Diabetes Prevention Program Research Group: Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med 346:393–403, 2002 [PMC free article: PMC1370926] [PubMed: 11832527]
220.
Perreault L, Pan Q, Mather KJ, Watson KE, Hamman RF, Kahn SE; Diabetes Prevention Program Research Group: Effect of regression from prediabetes to normal glucose regulation on long-term reduction in diabetes risk: results from the Diabetes Prevention Program Outcomes Study. Lancet 379:2243–2251, 2012 [PMC free article: PMC3555407] [PubMed: 22683134]
221.
Diabetes Prevention Program Outcomes Study Research Group; Orchard TJ, Temprosa M, Barrett-Connor E, Fowler SE, Goldberg RB, Mather KJ, Marcovina SM, Montez M, Ratner RE, Saudek CD, Sherif H, Watson KE: Long-term effects of the Diabetes Prevention Program interventions on cardiovascular risk factors: a report from the DPP Outcomes Study. Diabet Med 30:46–55, 2013 [PMC free article: PMC3524372] [PubMed: 22812594]
222.
Goldberg RB, Aroda VR, Bluemke DA, Barrett-Connor E, Budoff M, Crandall JP, Dabelea D, Horton ES, Mather KJ, Orchard TJ, Schade D, Watson K, Temprosa M; Diabetes Prevention Program Research Group: Effect of long-term metformin and lifestyle in the Diabetes Prevention Program and its outcome study on coronary artery calcium. Circulation 136:52–64, 2017 [PMC free article: PMC5526695] [PubMed: 28476766]
223.
Look AHEAD Research Group; Wing RR, Bolin P, Brancati FL, Bray GA, Clark JM, Coday M, Crow RS, Curtis JM, Egan CM, Espeland MA, Evans M, Foreyt JP, Ghazarian S, Gregg EW, Harrison B, Hazuda HP, Hill JO, Horton ES, Hubbard VS, Jakicic JM, Jeffery RW, Johnson KC, Kahn SE, Kitabchi AE, Knowler WC, Lewis CE, Maschak-Carey BJ, Montez MG, Murillo A, Nathan DM, Patricio J, Peters A, Pi-Sunyer X, Pownall H, Reboussin D, Regensteiner JG, Rickman AD, Ryan DH, Safford M, Wadden TA, Wagenknecht LE, West DS, Williamson DF, Yanovski SZ: Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. N Engl J Med 369:145–154, 2013
224.
National Institutes of Health. Weight Loss Does Not Lower Heart Disease Risk From Type 2 Diabetes [article online], 2012. Available from https://www​.nih.gov/news-events​/news-releases​/weight-loss-does-not-lower-heart-disease-risk-type-2-diabetes. Accessed 21 Mar 2022
225.
Nissen SE, Wolski K: Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med 356:2457–2471, 2007 [PubMed: 17517853]
226.
Regulations.gov. Guidance for Industry: Diabetes Mellitus—Evaluating Cardiovascular Risk in New Antidiabetic Therapies to Treat Type 2 Diabetes [article online], 2008. Available from https://www​.regulations​.gov/document/FDA-2008-D-0118-0029. Accessed 10 Oct 2023
227.
Cefalu WT, Kaul S, Gerstein HC, Holman RR, Zinman B, Skyler JS, Green JB, Buse JB, Inzucchi SE, Leiter LA, Raz I, Rosenstock J, Riddle MC: Cardiovascular outcomes trials in type 2 diabetes: where do we go from here? Reflections from a Diabetes Care Editors’ Expert Forum. Diabetes Care 41:14–31, 2018 [PMC free article: PMC5741160] [PubMed: 29263194]
228.
Bhatt DL, Szarek M, Pitt B, Cannon CP, Leiter LA, McGuire DK, Lewis JB, Riddle MC, Inzucchi SE, Kosiborod MN, Cherney DZI, Dwyer JP, Scirica BM, Bailey CJ, Diaz R, Ray KK, Udell JA, Lopes RD, Lapuerta P, Steg PG; SCORED Investigators: Sotagliflozin in patients with diabetes and chronic kidney disease. N Engl J Med 384:129–139, 2021 [PubMed: 33200891]
229.
Bhatt DL, Szarek M, Steg PG, Cannon CP, Leiter LA, McGuire DK, Lewis JB, Riddle MC, Voors AA, Metra M, Lund LH, Komajda M, Testani JM, Wilcox CS, Ponikowski P, Lopes RD, Verma S, Lapuerta P, Pitt B; SOLOIST-WHF Trial Investigators: Sotagliflozin in patients with diabetes and recent worsening heart failure. N Engl J Med 384:117–128, 2021 [PubMed: 33200892]
230.
ClinicalTrials.gov. A Study of Tirzepatide (LY3298176) Compared With Dulaglutide on Major Cardiovascular Events in Participants With Type 2 Diabetes (SURPASS-CVOT) [article online]. Available from https:​//clinicaltrials​.gov/ct2/show/NCT04255433.
231.
Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE; EMPA-REG OUTCOME Investigators: Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 373:2117–2128, 2015 [PubMed: 26378978]
232.
Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, Shaw W, Law G, Desai M, Matthews DR; CANVAS Program Collaborative Group: Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med 377:644–657, 2017 [PubMed: 28605608]
233.
Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Silverman MG, Zelniker TA, Kuder JF, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Gause-Nilsson IAM, Fredriksson M, Johansson PA, Langkilde AM, Sabatine MS; DECLARE-TIMI 58 Investigators: Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med 380:347–357, 2019 [PubMed: 30415602]
234.
Cannon CP, Pratley R, Dagogo-Jack S, Mancuso J, Huyck S, Masiukiewicz U, Charbonnel B, Frederich R, Gallo S, Cosentino F, Shih WJ, Gantz I, Terra SG, Cherney DZI, McGuire DK; VERTIS CV Investigators: Cardiovascular outcomes with ertugliflozin in type 2 diabetes. N Engl J Med 383:1425–1435, 2020 [PubMed: 32966714]
235.
Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Furtado RHM, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Sabatine MS: 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 139:2022–2031, 2019 [PubMed: 30786725]
236.
McMurray JJV, Solomon SD, Inzucchi SE, Kober L, Kosiborod MN, Martinez FA, Ponikowski P, Sabatine MS, Anand IS, Belohlavek J, Bohm M, Chiang CE, Chopra VK, de Boer RA, Desai AS, Diez M, Drozdz J, Dukat A, Ge J, Howlett JG, Katova T, Kitakaze M, Ljungman CEA, Merkely B, Nicolau JC, O’Meara E, Petrie MC, Vinh PN, Schou M, Tereshchenko S, Verma S, Held C, DeMets DL, Docherty KF, Jhund PS, Bengtsson O, Sjostrand M, Langkilde AM, DAPA-HF Trial Committees and Investigators: Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med 381:1995–2008, 2019 [PubMed: 31535829]
237.
Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, Januzzi J, Verma S, Tsutsui H, Brueckmann M, Jamal W, Kimura K, Schnee J, Zeller C, Cotton D, Bocchi E, Bohm M, Choi DJ, Chopra V, Chuquiure E, Giannetti N, Janssens S, Zhang J, Gonzalez Juanatey JR, Kaul S, Brunner-La Rocca HP, Merkely B, Nicholls SJ, Perrone S, Pina I, Ponikowski P, Sattar N, Senni M, Seronde MF, Spinar J, Squire I, Taddei S, Wanner C, Zannad F; EMPEROR-Reduced Trial Investigators: Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med 383:1413–1424, 2020 [PubMed: 32865377]
238.
Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Bohm M, Brunner-La Rocca HP, Choi DJ, Chopra V, Chuquiure-Valenzuela E, Giannetti N, Gomez-Mesa JE, Janssens S, Januzzi JL, Gonzalez-Juanatey JR, Merkely B, Nicholls SJ, Perrone SV, Pina IL, Ponikowski P, Senni M, Sim D, Spinar J, Squire I, Taddei S, Tsutsui H, Verma S, Vinereanu D, Zhang J, Carson P, Lam CSP, Marx N, Zeller C, Sattar N, Jamal W, Schnaidt S, Schnee JM, Brueckmann M, Pocock SJ, Zannad F, Packer M; EMPEROR-Preserved Trial Investigators: Empagliflozin in heart failure with a preserved ejection fraction. N Engl J Med 385:1451–1461, 2021 [PubMed: 34449189]
239.
Solomon SD, McMurray JJV, Claggett B, de Boer RA, DeMets D, Hernandez AF, Inzucchi SE, Kosiborod MN, Lam CSP, Martinez F, Shah SJ, Desai AS, Jhund PS, Belohlavek J, Chiang CE, Borleffs CJW, Comin-Colet J, Dobreanu D, Drozdz J, Fang JC, Alcocer-Gamba MA, Al Habeeb W, Han Y, Cabrera Honorio JW, Janssens SP, Katova T, Kitakaze M, Merkely B, O’Meara E, Saraiva JFK, Tereshchenko SN, Thierer J, Vaduganathan M, Vardeny O, Verma S, Pham VN, Wilderang U, Zaozerska N, Bachus E, Lindholm D, Petersson M, Langkilde AM; DELIVER Trial Committees and Investigators: Dapagliflozin in heart failure with mildly reduced or preserved ejection fraction. N Engl J Med 387:1089–1098, 2022 [PubMed: 36027570]
240.
Pfeffer MA, Claggett B, Diaz R, Dickstein K, Gerstein HC, Kober LV, Lawson FC, Ping L, Wei X, Lewis EF, Maggioni AP, McMurray JJ, Probstfield JL, Riddle MC, Solomon SD, Tardif JC; ELIXA Investigators: Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med 373:2247–2257, 2015 [PubMed: 26630143]
241.
Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg WM, Stockner M, Zinman B, Bergenstal RM, Buse JB; LEADER Steering Committee; LEADER Trial Investigators: Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 375:311–322, 2016 [PMC free article: PMC4985288] [PubMed: 27295427]
242.
Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst AG, Pettersson J, Vilsboll T; SUSTAIN-6: Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 375:1834–1844, 2016 [PubMed: 27633186]
243.
Holman RR, Bethel MA, Mentz RJ, Thompson VP, Lokhnygina Y, Buse JB, Chan JC, Choi J, Gustavson SM, Iqbal N, Maggioni AP, Marso SP, Ohman P, Pagidipati NJ, Poulter N, Ramachandran A, Zinman B, Hernandez AF; EXSCEL Study Group: Effects of once-weekly exenatide on cardiovascular outcomes in type 2 diabetes. N Engl J Med 377:1228–1239, 2017 [PMC free article: PMC9792409] [PubMed: 28910237]
244.
Hernandez AF, Green JB, Janmohamed S, D’Agostino RB, Sr., Granger CB, Jones NP, Leiter LA, Rosenberg AE, Sigmon KN, Somerville MC, Thorpe KM, McMurray JJV, Del Prato S; 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 392:1519–1529, 2018 [PubMed: 30291013]
245.
Gerstein HC, Colhoun HM, Dagenais GR, Diaz R, Lakshmanan M, Pais P, Probstfield J, Riesmeyer JS, Riddle MC, Ryden L, Xavier D, Atisso CM, Dyal L, Hall S, Rao-Melacini P, Wong G, Avezum A, Basile J, Chung N, Conget I, Cushman WC, Franek E, Hancu N, Hanefeld M, Holt S, Jansky P, Keltai M, Lanas F, Leiter LA, Lopez-Jaramillo P, Cardona Munoz EG, Pirags V, Pogosova N, Raubenheimer PJ, Shaw JE, Sheu WH, Temelkova-Kurktschiev T; REWIND Investigators: Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double-blind, randomised placebo-controlled trial. Lancet 394:121–130, 2019 [PubMed: 31189511]
246.
Husain M, Birkenfeld AL, Donsmark M, Dungan K, Eliaschewitz FG, Franco DR, Jeppesen OK, Lingvay I, Mosenzon O, Pedersen SD, Tack CJ, Thomsen M, Vilsboll T, Warren ML, Bain SC; PIONEER 6 Investigators: Oral semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 381:841–851, 2019 [PubMed: 31185157]
247.
Gerstein HC, Sattar N, Rosenstock J, Ramasundarahettige C, Pratley R, Lopes RD, Lam CSP, Khurmi NS, Heenan L, Del Prato S, Dyal L, Branch K; AMPLITUDE-O Trial Investigators: Cardiovascular and renal outcomes with efpeglenatide in type 2 diabetes. N Engl J Med 385:896–907, 2021 [PubMed: 34215025]
248.
Kristensen SL, Rorth R, Jhund PS, Docherty KF, Sattar N, Preiss D, Kober L, Petrie MC, McMurray JJV: Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet Diabetes Endocrinol 7:776–785, 2019 [PubMed: 31422062]
249.
Sattar N, Lee MMY, Kristensen SL, Branch KRH, Del Prato S, Khurmi NS, Lam CSP, Lopes RD, McMurray JJV, Pratley RE, Rosenstock J, Gerstein HC: Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a systematic review and meta-analysis of randomised trials. Lancet Diabetes Endocrinol 9:653–662, 2021 [PubMed: 34425083]
250.
Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Furtado RHM, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Sabatine MS: 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 393:31–39, 2019 [PubMed: 30424892]
251.
Patorno E, Htoo PT, Everett BM, Kim SC: Sodium-glucose cotransporter-2 inhibitors versus glucagon-like peptide-1 receptor agonists and the risk for cardiovascular outcomes in routine care patients with diabetes across categories of cardiovascular disease. Ann Intern Med 175:W4, 2022 doi:10.7326/L21-0708 [PubMed: 35038405] [CrossRef]
252.
Patient-Centered Outcomes Research Institute. PRECIDENTD (Prevention of Cardiovascular and Diabetic Kidney Disease in Type 2 Diabetes) [article online]. Available from https://www​.pcori.org​/research-results/2021​/precidentd-prevention-cardiovascular-and-diabetic-kidney-disease-type-2-diabetes.
253.
United States Renal Data System: 2018 USRDS Annual Data Report: Epidemiology of Kidney Disease in the United States. Bethesda, MD: National Institutes of Health, National Institute of Diabetes and Digestive and Kidney Diseases, 2018. Available from https://www​.niddk.nih​.gov/about-niddk/strategic-plans-reports​/usrds/prior-data-reports/2018
254.
Kidney Disease: Improving Global Outcomes (KDIGO) Diabetes Work Group: KDIGO 2020 clinical practice guideline for diabetes management in chronic kidney disease. Kidney Int 98:S1–S115, 2020 [PubMed: 32998798]
255.
Ninomiya T, Perkovic V, de Galan BE, Zoungas S, Pillai A, Jardine M, Patel A, Cass A, Neal B, Poulter N, Mogensen CE, Cooper M, Marre M, Williams B, Hamet P, Mancia G, Woodward M, Macmahon S, Chalmers J; ADVANCE Collaborative Group: Albuminuria and kidney function independently predict cardiovascular and renal outcomes in diabetes. J Am Soc Nephrol 20:1813–1821, 2009 [PMC free article: PMC2723977] [PubMed: 19443635]
256.
Fox CS, Matsushita K, Woodward M, Bilo HJ, Chalmers J, Heerspink HJ, Lee BJ, Perkins RM, Rossing P, Sairenchi T, Tonelli M, Vassalotti JA, Yamagishi K, Coresh J, de Jong PE, Wen CP, Nelson RG; Chronic Kidney Disease Prognosis Consortium: Associations of kidney disease measures with mortality and end-stage renal disease in individuals with and without diabetes: a meta-analysis. Lancet 380:1662–1673, 2012 [PMC free article: PMC3771350] [PubMed: 23013602]
257.
Arnett DK, Blumenthal RS, Albert MA, Buroker AB, Goldberger ZD, Hahn EJ, Himmelfarb CD, Khera A, Lloyd-Jones D, McEvoy JW, Michos ED, Miedema MD, Munoz D, Smith SC, Jr., Virani SS, Williams KA, Sr., Yeboah J, Ziaeian B: 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 140:e596–e646, 2019 [PMC free article: PMC7734661] [PubMed: 30879355]
258.
Afkarian M, Sachs MC, Kestenbaum B, Hirsch IB, Tuttle KR, Himmelfarb J, de Boer IH: Kidney disease and increased mortality risk in type 2 diabetes. J Am Soc Nephrol 24:302–308, 2013 [PMC free article: PMC3559486] [PubMed: 23362314]
259.
Sica DA, Bakris GL: Type 2 diabetes: RENAAL and IDNT—the emergence of new treatment options. J Clin Hypertens (Greenwich) 4:52–57, 2002 [PMC free article: PMC8099376] [PubMed: 11821641]
260.
McGuire DK, Shih WJ, Cosentino F, Charbonnel B, Cherney DZI, Dagogo-Jack S, Pratley R, Greenberg M, Wang S, Huyck S, Gantz I, Terra SG, Masiukiewicz U, Cannon CP: Association of SGLT2 inhibitors with cardiovascular and kidney outcomes in patients with type 2 diabetes: a meta-analysis. JAMA Cardiol 6:148–158, 2021 [PMC free article: PMC7542529] [PubMed: 33031522]
261.
Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, Edwards R, Agarwal R, Bakris G, Bull S, Cannon CP, Capuano G, Chu PL, de Zeeuw D, Greene T, Levin A, Pollock C, Wheeler DC, Yavin Y, Zhang H, Zinman B, Meininger G, Brenner BM, Mahaffey KW; CREDENCE Trial Investigators: Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med 380:2295–2306, 2019 [PubMed: 30990260]
262.
Heerspink HJL, Stefansson BV, Correa-Rotter R, Chertow GM, Greene T, Hou FF, Mann JFE, McMurray JJV, Lindberg M, Rossing P, Sjostrom CD, Toto RD, Langkilde AM, Wheeler DC; DAPA-CKD Committees and Investigators: Dapagliflozin in patients with chronic kidney disease. N Engl J Med 383:1436–1446, 2020 [PubMed: 32970396]
263.
EMPA-KIDNEY Collaborative Group; Herrington WG, Staplin N, Wanner C, Green JB, Hauske SJ, Emberson JR, Preiss D, Judge P, Mayne KJ, Ng SYA, Sammons E, Zhu D, Hill M, Stevens W, Wallendszus K, Brenner S, Cheung AK, Liu ZH, Li J, Hooi LS, Liu W, Kadowaki T, Nangaku M, Levin A, Cherney D, Maggioni AP, Pontremoli R, Deo R, Goto S, Rossello X, Tuttle KR, Steubl D, Petrini M, Massey D, Eilbracht J, Brueckmann M, Landray MJ, Baigent C, Haynes R: Empagliflozin in patients with chronic kidney disease. N Engl J Med 388:117–127, 2022
264.
Michos ED, Tuttle KR: GLP-1 receptor agonists in diabetic kidney disease. Clin J Am Soc Nephrol 16:1578–1580, 2021 [PMC free article: PMC8499005] [PubMed: 33849933]
265.
Bakris GL, Agarwal R, Anker SD, Pitt B, Ruilope LM, Rossing P, Kolkhof P, Nowack C, Schloemer P, Joseph A, Filippatos G; FIDELIO-DKD Investigators: Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. N Engl J Med 383:2219–2229, 2020 [PubMed: 33264825]
266.
Filippatos G, Anker SD, Agarwal R, Pitt B, Ruilope LM, Rossing P, Kolkhof P, Schloemer P, Tornus I, Joseph A, Bakris GL; FIDELIO-DKD Investigators: Finerenone and cardiovascular outcomes in patients with chronic kidney disease and type 2 diabetes. Circulation 143:540–552, 2021 [PMC free article: PMC7864612] [PubMed: 33198491]
267.
Agarwal R, Filippatos G, Pitt B, Anker SD, Rossing P, Joseph A, Kolkhof P, Nowack C, Gebel M, Ruilope LM, Bakris GL; 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 43:474–484, 2022 [PMC free article: PMC8830527] [PubMed: 35023547]
268.
Davies MJ, Aroda VR, Collins BS, Gabbay RA, Green J, Maruthur NM, Rosas SE, Del Prato S, Mathieu C, Mingrone G, Rossing P, Tankova T, Tsapas A, Buse JB: Management of hyperglycemia in type 2 diabetes, 2022. A Consensus Report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 45:2753–2786, 2022 [PMC free article: PMC10008140] [PubMed: 36148880]
269.
Blonde L, Umpierrez GE, Reddy SS, McGill JB, Berga SL, Bush M, Chandrasekaran S, DeFronzo RA, Einhorn D, Galindo RJ, Gardner TW, Garg R, Garvey WT, Hirsch IB, Hurley DL, Izuora K, Kosiborod M, Olson D, Patel SB, Pop-Busui R, Sadhu AR, Samson SL, Stec C, Tamborlane WV, Jr., Tuttle KR, Twining C, Vella A, Vellanki P, Weber SL: American Association of Clinical Endocrinology clinical practice guideline: developing a diabetes mellitus comprehensive care plan—2022 update. Endocr Pract 28:923–1049, 2022 [PMC free article: PMC10200071] [PubMed: 35963508]
270.
Das SR, Everett BM, Birtcher KK, Brown JM, Januzzi JL, Jr., Kalyani RR, Kosiborod M, Magwire M, Morris PB, Neumiller JJ, Sperling LS: 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 76:1117–1145, 2020 [PMC free article: PMC7545583] [PubMed: 32771263]
271.
ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Hilliard ME, Isaacs D, Johnson EL, Kahan S, Khunti K, Leon J, Lyons SK, Perry ML, Prahalad P, Pratley RE, Seley JJ, Stanton RC, Gabbay RA: 6. Glycemic targets: standards of care in diabetes—2023. Diabetes Care 46:S97–S110, 2023 [PMC free article: PMC9810469] [PubMed: 36507646]
272.
Cardoso R, Dudum R, Ferraro RA, Bittencourt M, Blankstein R, Blaha MJ, Nasir K, Rajagopalan S, Michos ED, Blumenthal RS, Cainzos-Achirica M: Cardiac computed tomography for personalized management of patients with type 2 diabetes mellitus. Circ Cardiovasc Imaging 13:e011365, 2020 [PubMed: 32900225]
273.
Orringer CE, Blaha MJ, Blankstein R, Budoff MJ, Goldberg RB, Gill EA, Maki KC, Mehta L, Jacobson TA: The National Lipid Association scientific statement on coronary artery calcium scoring to guide preventive strategies for ASCVD risk reduction. J Clin Lipidol 15:33–60, 2021 [PubMed: 33419719]
274.
Madaj PM, Budoff MJ, Li D, Tayek JA, Karlsberg RP, Karpman HL: Identification of noncalcified plaque in young persons with diabetes: an opportunity for early primary prevention of coronary artery disease identified with low-dose coronary computed tomographic angiography. Acad Radiol 19:889–893, 2012 [PMC free article: PMC4277701] [PubMed: 22542200]
275.
Leipsic J, Taylor CM, Gransar H, Shaw LJ, Ahmadi A, Thompson A, Humphries K, Berman DS, Hausleiter J, Achenbach S, Al-Mallah M, Budoff MJ, Cademartiri F, Callister TQ, Chang HJ, Chow BJ, Cury RC, Delago AJ, Dunning AL, Feuchtner GM, Hadamitzky M, Kaufmann PA, Lin FY, Chinnaiyan KM, Maffei E, Raff GL, Villines TC, Gomez MJ, Min JK: Sex-based prognostic implications of nonobstructive coronary artery disease: results from the international multicenter CONFIRM study. Radiology 273:393–400, 2014 [PMC free article: PMC4334228] [PubMed: 25028784]
276.
Ferencik M, Mayrhofer T, Bittner DO, Emami H, Puchner SB, Lu MT, Meyersohn NM, Ivanov AV, Adami EC, Patel MR, Mark DB, Udelson JE, Lee KL, Douglas PS, Hoffmann U: Use of high-risk coronary atherosclerotic plaque detection for risk stratification of patients with stable chest pain: a secondary analysis of the PROMISE randomized clinical trial. JAMA Cardiol 3:144–152, 2018 [PMC free article: PMC5838601] [PubMed: 29322167]
277.
Ferraro R, Latina JM, Alfaddagh A, Michos ED, Blaha MJ, Jones SR, Sharma G, Trost JC, Boden WE, Weintraub WS, Lima JAC, Blumenthal RS, Fuster V, Arbab-Zadeh A: Evaluation and management of patients with stable angina: beyond the ischemia paradigm: JACC State-of-the-Art Review. J Am Coll Cardiol 76:2252–2266, 2020 [PubMed: 33153586]
278.
Gulati M, Levy PD, Mukherjee D, Amsterdam E, Bhatt DL, Birtcher KK, Blankstein R, Boyd J, Bullock-Palmer RP, Conejo T, Diercks DB, Gentile F, Greenwood JP, Hess EP, Hollenberg SM, Jaber WA, Jneid H, Joglar JA, Morrow DA, O’Connor RE, Ross MA, Shaw LJ: 2021 AHA/ACC/ASE/CHEST/SAEM/SCCT/SCMR guideline for the evaluation and diagnosis of chest pain: executive summary: a report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation 144:e368–e454, 2021 [PubMed: 34709928]
279.
He C, Yang ZG, Chu ZG, Dong ZH, Li YM, Shao H, Deng W: Comparison of carotid and cerebrovascular disease between diabetic and non-diabetic patients using dual-source CT. Eur J Radiol 80:e361–365, 2011 [PubMed: 21227617]
280.
Vigili de Kreutzenberg S, Fadini GP, Guzzinati S, Mazzucato M, Volpi A, Coracina A, Avogaro A: Carotid plaque calcification predicts future cardiovascular events in type 2 diabetes. Diabetes Care 38:1937–1944, 2015 [PubMed: 26253729]
281.
US Preventive Services Task Force; Davidson KW, Barry MJ, Mangione CM, Cabana M, Chelmow D, Coker TR, Davis EM, Donahue KE, Jaen CR, Krist AH, Kubik M, Li L, Ogedegbe G, Pbert L, Ruiz JM, Stevermer J, Tseng CW, Wong JB: Aspirin use to prevent cardiovascular disease: US Preventive Services Task Force Recommendation Statement. JAMA 327:1577–1584, 2022 [PubMed: 35471505]
282.
Stempniewicz N, Vassalotti JA, Cuddeback JK, Ciemins E, Storfer-Isser A, Sang Y, Matsushita K, Ballew SH, Chang AR, Levey AS, Bailey RA, Fishman J, Coresh J: Chronic kidney disease testing among primary care patients with type 2 diabetes across 24 U.S. health care organizations. Diabetes Care 44:2000–2009, 2021 [PMC free article: PMC8740923] [PubMed: 34233925]
283.
Centers for Medicare and Medicaid Services. Chronic Conditions Data Warehouse [article online]. Available from https://www2​.ccwdata.org/web/guest/home/.

Appendices

APPENDIX A1.

Crude and Age-Standardized Prevalence of History of Heart Disease Among Adults Age ≥18 Years, by Diabetes Status, Sex, and Race/Ethnicity, NHIS, U.S., 2019–2020

CHARACTERISTICSPERCENT (STANDARD ERROR)
DiabetesNo DiabetesDiabetesNo DiabetesDiabetesNo Diabetes
Coronary Heart DiseaseAngina PectorisHeart Attack
Total
Crude16.5 (0.63)3.4 (0.11)5.4 (0.37)1.2 (0.06)10.4 (0.50)2.3 (0.09)
Age-standardized16.5 (0.60)6.9 (0.20)5.5 (0.37)2.0 (0.10)10.4 (0.49)4.6 (0.17)
Sex
Men
Crude20.0 (1.00)4.4 (0.18)5.9 (0.59)1.2 (0.09)13.2 (0.80)3.1 (0.15)
Age-standardized20.0 (0.94)9.6 (0.36)5.9 (0.59)2.3 (0.15)13.2 (0.77)6.5 (0.31)
Women
Crude13.0 (0.73)2.4 (0.12)5.0 (0.45)1.1 (0.08)7.5 (0.56)1.6 (0.09)
Age-standardized12.9 (0.71)4.7 (0.23)5.0 (0.44)1.8 (0.14)7.5 (0.55)3.0 (0.18)
Race/ethnicity
Non-Hispanic White
Crude19.8 (0.83)4.0 (0.14)6.5 (0.49)1.3 (0.08)13.4 (0.71)2.9 (0.12)
Age-standardized18.8 (0.79)7.1 (0.23)6.3 (0.49)2.1 (0.12)12.8 (0.68)5.0 (0.20)
Non-Hispanic Black
Crude13.4 (1.35)2.8 (0.29)3.6 (0.71)1.0 (0.21)7.2 (0.95)1.9 (0.23)
Age-standardized14.0 (1.39)6.6 (0.74)3.9 (0.77)1.8 (0.31)7.5 (1.00)4.3 (0.60)
All Hispanic
Crude10.1 (1.20)1.8 (0.20)4.0 (0.95)0.8 (0.14)5.7 (0.93)0.8 (0.11)
Age-standardized12.2 (1.41)5.7 (0.66)4.5 (0.90)1.4 (0.24)6.9 (1.11)2.3 (0.37)
Mexican American
Crude9.3 (1.62)1.4 (0.22)3.0 (0.66)0.7 (0.16)5.0 (1.11)0.6 (0.12)
Age-standardized11.7 (1.89)5.7 (1.07)3.8 (0.80)1.4 (0.33)6.0 (1.29)2.5 (0.58)
Other Hispanic
Crude11.8 (2.03)2.4 (0.37)5.9 (2.38)0.8 (0.26)7.1 (1.78)0.9 (0.20)
Age-standardized13.5 (2.34)6.0 (0.86)6.1 (2.17)11.4 (0.38)8.4 (1.98)2.3 (0.51)
Non-Hispanic Asian
Crude10.2 (3.21)12.3 (0.39)2.2 (0.77)0.7 (0.15) 2 0.8 (0.26)1
Age-standardized9.9 (2.90)5.2 (1.02)2.3 (0.79)11.6 (0.37) 2 2.2 (0.77)1
Any Heart Condition, Including AnginaAny Heart Condition, not Including Angina
Total
Crude20.7 (0.67)4.8 (0.13)19.1 (0.67)4.3 (0.12)
Age-standardized20.6 (0.65)9.3 (0.23)19.1 (0.64)8.6 (0.22)
Sex
Men
Crude24.8 (1.10)5.9 (0.21)23.2 (1.07)5.6 (0.21)
Age-standardized24.8 (1.04)12.3 (0.39)23.2 (1.00)11.9 (0.39)
Women
Crude16.5 (0.79)3.7 (0.15)15.0 (0.78)3.2 (0.13)
Age-standardized16.5 (0.78)6.8 (0.27)15.0 (0.76)6.0 (0.25)
Race/ethnicity
Non-Hispanic White
Crude24.8 (0.89)5.6 (0.17)23.3 (0.90)5.2 (0.16)
Age-standardized23.6 (0.85)9.6 (0.26)22.1 (0.85)9.0 (0.25)
Non-Hispanic Black
Crude15.8 (1.43)4.4 (0.38)14.9 (1.39)3.8 (0.33)
Age-standardized16.6 (1.46)9.7 (0.87)15.5 (1.43)8.9 (0.86)
All Hispanic
Crude14.1 (1.45)2.6 (0.24)11.9 (1.26)2.1 (0.20)
Age-standardized16.9 (1.55)7.4 (0.74)14.5 (1.44)6.6 (0.70)
Mexican American
Crude12.2 (1.73)2.22 (0.27)10.9 (1.69)1.7 (0.24)
Age-standardized15.3 (1.98)7.3 (1.12)13.6 (1.94)6.5 (1.11)
Other Hispanic
Crude18.0 (2.81)3.2 (0.46)14.0 (2.16)2.8 (0.38)
Age-standardized20.1 (2.90)7.6 (1.06)16.2 (2.37)6.9 (0.93)
Non-Hispanic Asian
Crude10.6 (3.27)2.7 (0.41)10.2 (3.21)12.5 (0.41)
Age-standardized10.3 (2.98)6.1 (1.08)9.9 (2.90)5.5 (1.07)

History of heart disease and diabetes status are self-reported. Where noted, data are standardized to the NHIS 2019–2020 population with diabetes, using age categories 20–44, 45–64, and ≥65 years.

1

Relative standard error >30%–40%

2

Estimate is too unreliable to present; ≤1 case or relative standard error >50%.

SOURCE: National Health Interview Surveys (NHIS) 2019–2020

APPENDIX A2.

Crude and Age-Standardized Prevalence of History of Heart Disease Among Adults Age ≥20 Years, by Diabetes Status, Sex, and Race/Ethnicity, NHANES, U.S., 2017–2020 Q1

CHARACTERISTICSPERCENT (STANDARD ERROR)
DiabetesNo DiabetesDiabetesNo DiabetesDiabetesNo Diabetes
Congestive Heart FailureCoronary Heart DiseaseAngina
Total
Crude9.6 (1.19)1.7 (0.18)14.8 (2.56)2.9 (0.38)8.7 (1.19)1.5 (0.18)
Age-standardized9.7 (1.08)3.2 (0.32)15.0 (2.40)5.7 (0.58)8.7 (1.16)2.6 (0.36)
Sex
Men
Crude11.8 (2.09)1.8 (0.23)18.9 (3.30)4.1 (0.66)11.7 (2.11)1.8 (0.25)
Age-standardized11.6 (1.79)3.3 (0.44)19.1 (2.91)9.1 (1.14)11.4 (2.01)3.7 (0.55)
Women
Crude6.9 (1.07)1.7 (0.24)9.9 (2.50)1.7 (0.34)5.2 (1.17)1.2 (0.18)
Age-standardized7.2 (1.07)3.1 (0.37)10.3 (2.55)3.2 (0.59)5.4 (1.23)1.8 (0.34)
Race/ethnicity
Non-Hispanic White
Crude11.1 (1.93)1.9 (0.26)20.0 (4.21)3.7 (0.57)10.9 (1.83)1.8 (0.25)
Age-standardized10.9 (1.83)3.2 (0.39)19.7 (3.99)6.3 (0.77)10.8 (1.78)2.7 (0.42)
Non-Hispanic Black
Crude11.2 (2.04)2.6 (0.33)6.6 (1.31)1.1 (0.22)3.9 (0.88)0.9 (0.23)
Age-standardized11.7 (2.02)5.5 (0.79)6.8 (1.31)2.8 (0.52)4.0 (0.91)1.6 (0.39)
All Hispanic
Crude5.4 (1.06)0.8 (0.24)6.0 (1.35)1.4 (0.33)6.0 (1.31)0.8 (0.19)
Age-standardized6.9 (1.39)2.2 (0.63)7.5 (1.43)4.7 (1.16)6.6 (1.34)2.7 (0.86)1
Mexican American
Crude3.4 (1.45)20.5 (0.17)13.9 (1.35)11.3 (0.40)13.3 (1.58)20.6 (0.21)1
Age-standardized6.0 (2.42)22.8 (1.35)26.6 (2.15)16.9 (2.62)13.3 (1.42)23.5 (1.69)2
Other Hispanic
Crude8.1 (1.82)1.2 (0.48)28.9 (2.78)11.6 (0.47)19.8 (2.21)0.9 (0.30)1
Age-standardized8.7 (1.97)2.0 (0.79)19.9 (3.33)13.6 (1.02)10.3 (2.32)2.4 (0.86)1
Other/Multiracial
Crude5.2 (1.36)1.3 (0.43)18.1 (1.77)1.8 (0.47)6.1 (1.67)1.5 (0.64)2
Age-standardized5.9 (1.66)2.7 (0.94)18.3 (1.90)5.1 (1.38)6.0 (1.78)3.2 (1.38)2
Heart AttackAny Heart Condition, Including AnginaAny Heart Condition, not Including Angina
Total
Crude12.8 (1.81)2.6 (0.37)23.9 (2.72)5.5 (0.51)22.2 (2.77)4.9 (0.52)
Age-standardized12.8 (1.66)4.8 (0.58)24.2 (2.52)10.3 (0.76)22.5 (2.57)9.5 (0.75)
Sex
Men
Crude15.3 (2.34)3.8 (0.60)29.0 (3.29)6.6 (0.67)26.9 (3.49)6.1 (0.77)
Age-standardized15.2 (2.18)7.9 (0.94)29.0 (2.78)13.9 (1.02)26.9 (2.98)13.1 (1.12)
Women
Crude10.0 (2.25)1.5 (0.30)17.9 (2.97)4.4 (0.58)16.7 (2.84)3.7 (0.49)
Age-standardized10.3 (2.23)2.5 (0.52)18.6 (2.98)7.7 (0.96)17.4 (2.87)6.7 (0.84)
Race/ethnicity
Non-Hispanic White
Crude15.6 (2.84)3.0 (0.51)28.2 (4.45)6.5 (0.73)26.8 (4.53)5.8 (0.74)
Age-standardized15.4 (2.72)4.8 (0.72)27.8 (4.19)10.7 (0.99)26.4 (4.28)9.7 (0.98)
Non-Hispanic Black
Crude10.6 (1.92)2.0 (0.33)18.3 (2.66)4.6 (0.53)17.8 (2.61)4.2 (0.45)
Age-standardized10.9 (1.94)4.1 (0.61)18.9 (2.58)9.8 (0.98)18.4 (2.56)9.2 (0.88)
All Hispanic
Crude6.8 (1.85)1.3 (0.28)15.0 (2.80)2.6 (0.43)11.5 (2.18)2.4 (0.39)
Age-standardized7.7 (1.95)4.3 (1.00)17.4 (2.63)8.7 (1.67)13.9 (2.20)7.8 (1.47)
Mexican American
Crude3.1 (1.39)20.8 (0.19)10.4 (3.44)12.1 (0.48)7.8 (2.48)11.8 (0.40)
Age-standardized4.3 (1.61)14.5 (1.28)14.4 (4.06)11.3 (3.53)112.2 (3.26)9.9 (2.89)
Other Hispanic
Crude11.8 (3.20)1.7 (0.47)21.4 (4.23)3.2 (0.58)16.6 (3.48)2.9 (0.54)
Age-standardized12.5 (3.68)4.3 (1.26)22.4 (4.52)7.6 (1.33)17.7 (4.11)7.0 (1.27)
Other/Multiracial
Crude9.3 (3.54)12.6 (0.62)19.5 (3.46)4.4 (0.88)17.3 (3.43)3.7 (0.79)
Age-standardized8.7 (2.71)16.9 (1.71)19.3 (2.80)10.2 (1.88)17.3 (2.76)9.4 (1.94)

History of heart disease and diabetes status are self-reported. Where noted, data are standardized to the National Health Interview Surveys 2019–2020 population with diabetes, using age categories 20–44, 45–64, and ≥65 years.

1

Relative standard error >30%–40%

2

Relative standard error >40%–50%

SOURCE: National Health and Nutrition Examination Surveys (NHANES) 2017–2020 Q1

APPENDIX A3.

Crude and Age-Standardized Prevalence of History of Heart Disease Among Adults Age ≥18 Years, by Diabetes Status, Sex, Race/Ethnicity, and Other Characteristics, BRFSS, U.S., 2019

CHARACTERISTICSPERCENT (STANDARD ERROR)
DiabetesNo DiabetesDiabetesNo Diabetes
Coronary Heart Disease or AnginaHeart Attack or Myocardial Infarction
Total
Crude12.7 (0.28)2.8 (0.05)13.5 (0.29)3.1 (0.05)
Age-standardized12.7 (0.27)5.2 (0.08)13.5 (0.29)5.5 (0.08)
Sex
Men
Crude14.7 (0.43)3.4 (0.08)17.0 (0.47)3.9 (0.08)
Age-standardized14.7 (0.42)7.0 (0.15)17.0 (0.47)7.7 (0.15)
Women
Crude10.7 (0.35)2.2 (0.05)10.0 (0.34)2.3 (0.06)
Age-standardized10.7 (0.35)3.8 (0.09)10.0 (0.34)3.7 (0.09)
Race/ethnicity
Non-Hispanic White
Crude14.6 (0.32)3.4 (0.06)15.2 (0.34)3.5 (0.06)
Age-standardized13.9 (0.31)5.5 (0.09)14.7 (0.34)5.6 (0.09)
Non-Hispanic Black
Crude9.6 (0.61)2.1 (0.14)10.6 (0.65)2.4 (0.14)
Age-standardized10.0 (0.64)4.2 (0.30)10.7 (0.63)5.0 (0.29)
All Hispanic
Crude8.3 (0.70)1.4 (0.09)10.7 (0.86)2.1 (0.14)
Age-standardized9.4 (0.79)3.3 (0.29)11.4 (0.88)4.9 (0.41)
Non-Hispanic Asian
Crude14.9 (3.12)1.2 (0.27)9.1 (2.37)1.3 (0.23)
Age-standardized16.3 (3.22)3.1 (0.72)9.8 (2.43)2.7 (0.58)
Native Hawaiian/Other Pacific Islander
Crude 3 2.0 (0.54)18.6 (7.88)23.1 (0.69)
Age-standardized16.0 (6.69)24.0 (1.33)118.3 (6.59)16.7 (1.97)
American Indian/Alaska Native
Crude15.4 (2.22)5.1 (0.73)18.0 (2.24)6.3 (0.60)
Age-standardized16.7 (2.39)8.3 (1.07)19.6 (2.35)9.8 (0.98)
BMI, kg/m2
<25
Crude12.8 (0.74)2.3 (0.09)13.5 (0.75)2.6 (0.08)
Age-standardized12.0 (0.72)4.4 (0.13)13.0 (0.76)5.0 (0.14)
25–29.9
Crude13.1 (0.53)3.2 (0.09)14.4 (0.58)3.3 (0.09)
Age-standardized12.3 (0.51)5.6 (0.15)13.8 (0.58)5.6 (0.15)
≥30
Crude12.8 (0.39)3.4 (0.09)13.3 (0.40)3.6 (0.10)
Age-standardized13.5 (0.39)6.4 (0.18)13.7 (0.40)6.4 (0.18)
Smoking status
Current
Crude15.8 (1.00)3.3 (0.12)18.7 (1.11)4.8 (0.17)
Age-standardized17.5 (1.09)6.6 (0.27)19.6 (1.05)9.2 (0.33)
Former
Crude16.7 (0.49)9.3 (0.34)17.5 (0.53)5.5 (0.14)
Age-standardized15.4 (0.45)7.0 (0.17)16.5 (0.53)7.3 (0.18)
Never
Crude9.3 (0.34)1.8 (0.05)9.6 (0.32)1.8 (0.05)
Age-standardized9.5 (0.34)3.8 (0.10)9.7 (0.32)3.6 (0.09)
Heavy drinker
Crude10.4 (0.40)2.3 (0.06)11.0 (0.43)2.4 (0.06)
Age-standardized11.0 (0.41)4.7 (0.11)11.4 (0.43)4.7 (0.11)
High blood pressure
Crude15.5 (0.36)7.2 (0.15)16.0 (0.36)7.4 (0.14)
Age-standardized14.9 (0.34)8.1 (0.15)15.6 (0.37)8.3 (0.15)
High cholesterol
Crude16.2 (0.41)7.4 (0.16)16.4 (0.41)7.2 (0.15)
Age-standardized15.8 (0.39)8.6 (0.17)16.1 (0.41)8.2 (0.16)
Preventive aspirin use
Crude18.1 (1.18)10.5 (0.58)20.3 (1.55)12.7 (0.69)
Age-standardized16.4 (1.07)10.2 (0.57)20.2 (1.99)12.3 (0.69)
Any physical activity
Crude11.6 (0.38)2.5 (0.06)12.2 (0.39)2.6 (0.05)
Age-standardized11.8 (0.38)4.8 (0.09)12.3 (0.39)4.8 (0.10)

All data are self-reported. Where noted, data are standardized to the BFRSS 2019 population with diabetes, using age categories 20–44, 45–64, and ≥65 years.

1

Relative standard error >30%–40%

2

Relative standard error >40%–50%

3

Estimate is too unreliable to present; ≤1 case or relative standard error >50%.

SOURCE: Behavioral Risk Factor Surveillance System (BRFSS) 2019

APPENDIX A4.

Crude and Age-Standardized Prevalence of History of Heart Disease Among Adults Age ≥18 Years, by Diabetes Status, Sex, and Race, HCUP-NIS, U.S., 2018

CHARACTERISTICSPERCENT (STANDARD ERROR)
DiabetesNo DiabetesDiabetesNo DiabetesDiabetesNo Diabetes
Myocardial InfarctionAnginaCardiac Dysrhythmia
Total
Crude5.8 (0.02)3.0 (0.01)0.1 (<0.01)0.1 (<0.01)15.5 (0.03)10.7 (0.02)
Age-standardized5.6 (0.02)3.8 (0.01)0.1 (<0.01)0.1 (<0.01)14.4 (0.03)13.2 (0.02)
Sex
Men
Crude6.6 (0.03)4.5 (0.02)0.1 (<0.01)0.1 (<0.01)17.4 (0.04)14.3 (0.03)
Age-standardized6.4 (0.03)4.9 (0.02)0.1 (<0.01)0.1 (<0.01)16.2 (0.04)15.5 (0.03)
Women
Crude4.9 (0.02)2.0 (0.01)0.2 (<0.01)0.1 (<0.01)13.6 (0.04)8.3 (0.02)
Age-standardized4.7 (0.02)2.9 (0.01)0.2 (<0.01)0.1 (<0.01)12.4 (0.04)11.3 (0.02)
Race
White
Crude6.1 (0.02)3.3 (0.01)0.1 (<0.01)0.1 (<0.01)17.1 90.04)12.2 (0.02)
Age-standardized5.8 (0.02)3.9 (0.01)0.1 (<0.01)0.1 (<0.01)15.1 (0.03)13.4 (0.02)
Black
Crude4.6 (0.04)2.5 (0.02)0.2 (<0.01)0.1 (<0.01)13.2 (0.06)8.5 (0.04)
Age-standardized4.7 (0.04)3.7 (0.03)0.2 (<0.01)0.2 (<0.01)13.6 (0.06)13.1 (0.06)
Other
Crude5.9 (0.04)2.2 (0.02)0.2 (<0.01)0.1 (<0.01)12.8 (0.06)7.0 (0.03)
Age-standardized5.8 (0.04)3.9 (0.03)0.2 (<0.01)0.1 (<0.01)12.4 (0.06)11.9 (0.05)
Congestive Heart FailureCoronary Heart DiseaseCardiovascular Disease
Total
Crude31.2 (0.04)13.0 (0.02)37.7 (0.04)16.7 (0.02)57.2 (0.04)31.3 (0.02)
Age-standardized29.5 (0.04)16.3 (0.02)35.6 (0.04)21.4 (0.02)54.4 (0.04)39.1 (0.03)
Sex
Men
Crude31.9 (0.05)16.4 (0.03)43.5 (0.05)24.8 (0.03)61.1 (0.05)41.2 (0.04)
Age-standardized30.4 (0.05)17.8 (0.03)41.0 (0.05)27.3 (0.03)58.2 (0.05)44.7 (0.04)
Women
Crude30.3 (0.05)10.7 (0.02)31.8 (0.05)11.5 (0.02)53.2 (0.06)24.8 (0.03)
Age-standardized28.6 (0.05)14.9 (0.03)30.0 (0.05)16.3 (0.03)50.5 (0.05)34.4 (0.03)
Race
White
Crude31.8 (0.05)14.1 (0.02)41.1 (0.05)19.4 (0.02)60.0 (0.05)35.1 (0.03)
Age-standardized28.8 (0.04)15.6 (0.02)37.6 (0.05)21.9 (0.03)55.3 (0.05)39.2 (0.03)
Black
Crude34.1 (0.09)14.5 (0.05)31.1 (0.09)12.9 (0.04)54.6 (0.09)27.9 (0.06)
Age-standardized34.5 (0.09)22.1 (0.07)31.6 (0.09)20.6 (0.07)55.4 (0.09)41.8 (0.08)
Other
Crude26.8 (0.08)8.1 (0.03)33.3 (0.08)10.5 (0.03)51.0 (0.09)20.8 (0.04)
Age-standardized26.3 (0.07)14.2 (0.05)32.7 (0.08)18.9 (0.06)50.2 (0.08)35.4 (0.07)

Heart disease and diabetes are defined by International Classification of Diseases, Tenth Revision (ICD-10) codes based on the Centers for Medicare and Medicaid Services (CMS) Chronic Condition Data Warehouse (CCW) Condition Categories (Reference 283). Where noted, data are standardized to the National Health Interview Surveys 2019–2020 population with diabetes, using age categories 20–44, 45–64, and ≥65 years.

SOURCE: Healthcare Cost and Utilization Project–National Inpatient Sample (HCUP-NIS) 2018

Dr. Kalyani is a standing member of the FDA Endocrinologic and Metabolic Drugs Advisory Committee. Dr. Everett has received consulting fees from Circulation (Associate Editor), Eli Lilly & Co, Gilead Sciences, Ipsen Biosciences Inc, Janssen (Johnson & Johnson), Provention Bio, and UpToDate (royalties for peer review content). Dr. Everett is a co-investigator on a grant funded by Novo Nordisk, which is paid to his institution. Dr. Perrault reported no conflicts of interest. Dr. Michos has received consulting fees from AstraZeneca, Amarin, Amgen, Bayer, Boehringer Ingelheim, Edwards Lifesciences, Esperion Therapeutics, Medtronic, Novartis, Novo Nordisk, and Pfizer. Dr. Michos is a co-investigator on a grant funded by Merck, which is paid to her institution.

Copyright Notice

Diabetes in America is in the public domain of the United States. You may use the work without restriction in the United States.

Bookshelf ID: NBK597416PMID: 38117924

Views

  • PubReader
  • Print View
  • Cite this Page

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Similar articles in PubMed

See reviews...See all...

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...