Econstudentlog

A few diabetes papers of interest

i. Glycemic Control and Risk of Infections Among People With Type 1 or Type 2 Diabetes in a Large Primary Care Cohort Study. From the paper:

“Infections are widely considered to be a source of significant health care costs and to reduce quality of life among people with diabetes mellitus (DM) (1). Nevertheless, relatively few, large, well-designed, epidemiological studies have explored relationships between poorer control of DM and infections; previous studies have important limitations (1). Most randomized controlled trials (RCTs) of DM control have not investigated the effect of improved glycemic control on infections and are unlikely to do so at present because of the high cost and lack of good-quality supporting observational evidence. […] A recent review of higher-quality population-based epidemiological studies found clinically important (∼1.5–3.5 times higher) infection risks associated with poorer DM control in some studies (usually defined as a glycated hemoglobin [HbA1c] level >7–8% [53–64 mmol/mol]) (1). However, the studies were inconsistent, generating uncertainty about the evidence.

A key concern with previous work is that the measurement of HbA1c usually was made at or near to the time of the infection, so any association could be explained by reverse causality. Any infectious disease episode can itself have an adverse effect on glycemic control, a process known as stress hyperglycemia (4); hence, blood glucose or HbA1c measurements near the time of an infection may be elevated, rendering determination of the chronology and relationship between the two difficult. Several studies with serial HbA1c measurements have shown that the stress hyperglycemia response can be substantial (46). Another important issue is that studies of incident DM often use measurements of HbA1c obtained during initial presentation, and these typically do not represent subsequent levels after initiation of treatment; use of such measurements may obscure associations between usual HbA1c level and infection risk. Other limitations of previous work include a lack of consideration of type of DM (especially T1DM) and fewer older people with DM. The current study uses a large English primary care database with repeated HbA1c measurements wherein we can classify individuals more precisely in terms of their baseline glycemic control as well as ensure that these HbA1c measurements were made before the infection episode.”

“With the use of English primary care data, average glycated hemoglobin (HbA1c) during 2008–2009 was estimated for 85,312 patients with DM ages 40–89 years. Infection rates during 2010–2015 compiled from primary care, linked hospital, and mortality records were estimated across 18 infection categories and further summarized as any requiring a prescription or hospitalization or as cause of death. Poisson regression was used to estimate adjusted incidence rate ratios (IRRs) by HbA1c categories across all DM, and type 1 and type 2 DM separately. IRRs also were compared with 153,341 age-sex-practice–matched controls without DM. Attributable fractions (AF%) among patients with DM were estimated for an optimal control scenario (HbA1c 6–7% [42–53 mmol/mol]).”

“Crude infection rates during 2010–2015 estimated across 18 different categories confirmed consistently higher rates among patients with DM […]. Long-term infection risk rose with increasing HbA1c for most outcomes. Compared with patients without DM, those with DM and optimal control (HbA1c 6–7% [42–53 mmol/mol], IRR 1.41 [95% CI 1.36–1.47]) and poor control (≥11% [97 mmol/mol], 4.70 [4.24–5.21]) had elevated hospitalization risks for infection. In patients with type 1 DM and poor control, this risk was even greater (IRR 8.47 [5.86–12.24]). Comparisons within patients with DM confirmed the risk of hospitalization with poor control (2.70 [2.43–3.00]) after adjustment for duration and other confounders. AF% of poor control were high for serious infections, particularly bone and joint (46%), endocarditis (26%), tuberculosis (24%), sepsis (21%), infection-related hospitalization (17%), and mortality (16%). […] even patients with DM with good control were at an increased risk compared with matched controls without DM. Thus, compared with patients without DM, patients with DM and good control (mean HbA1c 6–7%, IRR 1.41 [95% CI 1.36–1.47]) and those with poor control (≥11%, 4.70 [4.24–5.21]) had elevated hospitalization risks for infection. These risks were higher among patients with T1DM. For example, patients with T1DM with a mean HbA1c ≥11%, had more than eight times the risk of hospitalization than their matched controls without DM (IRR 8.47 [5.86–12.24]), whereas for T2DM, this was four times higher (4.31 [3.88–4.80]). […] Patients with T1DM […] had higher rates of hospitalization (1.12 [1.01–1.24]) and death as a result of infection (1.42 [1.03–1.96]) than patients with T2DM, even after accounting for duration of DM.”

“In terms of the overall population effect, almost one-half of bone and joint infections among patients with DM were attributed to poor control. […] The most novel and concerning finding is the substantial proportion of other serious infections statistically attributable to poor glycemic control, particularly endocarditis, tuberculosis, and sepsis. Between 20 and 30% of these infections in the English DM population could be attributed to poor control. […] [W]e estimated AF% for the three summary groupings […] plus individual infection types […] across HbA1c categories for patients with DM compared with the optimal control scenario of 6–7%. The largest AF% estimate was for bone and joint infections, with 46.0% of hospitalizations being attributed to HbA1c values outside of the range 6–7%. Other large AF% estimates were observed for endocarditis (26.2%) and tuberculosis (23.7%), but CIs were wide. Sepsis (20.8%), pneumonia (15.3%), skin infections (cellulitis 14.0%, other 12.1%), and candidiasis (16.5%) all produced AF% estimates of ≥10%. Overall, 15.7% of infection-related deaths, 16.5% of infection-related hospitalizations, and 6.8% of infections requiring a prescription were attributed to values of HbA1c outside the 6–7% range.”

“Prevalence of diagnosed T2DM has tripled in the U.K. over the past 20 years (17). Although some improvements in glycemic control also have been observed over this period, our analyses show that substantial numbers of patients still have very poor glycemic control (e.g., 16% of patients with T2DM and 41% of patients with T1DM had a mean HbA1c >9%). […] 14% of patients with DM in the current study were hospitalized for infection during follow-up […] The U.K. has a relatively low prevalence of DM and good control on the basis of international comparisons (18); therefore, in many low- and middle-income countries, the burden of infections attributable to poor glycemic control could be substantially higher (19).”

“A variety of mechanisms may link DM and hyperglycemia with infection response (1,2022). Diabetes progression itself is associated with immune dysfunction; autoimmunity in T1DM and low-grade chronic inflammation in T2DM (1). Hyperglycemia may also have adverse effects on several types of immune cells (19,23); alter cytokine and chemokine gene expression (24), and inhibit effects of complement (25). Other important mechanisms may include peripheral diabetic neuropathy because this results in a loss of sensation and reduced awareness of minor injuries (13). Alongside ischemia, often as a result of related peripheral arterial disease, neuropathy can result in impaired barrier defenses, skin ulcers, and lesions with poor wound healing and an increased risk of secondary infections (19). Although numerous mechanisms exist, nearly all involve poor glycemic control. Thus, that improved control would reduce infections seems likely […] Overall, the current analyses demonstrate a strong and likely causal association between hyperglycemia and infection risk for both T1DM and T2DM. DM duration and other markers of severity cannot explain the increased risk, nor can longer duration explain the increased risk for T1DM compared with T2DM. This remains the case in older people in whom infections are common and often severe and more uncertainty exists about the vascular benefits of improving DM control. Substantial proportions of serious infections can be attributed to poor control, even though DM is managed well in the U.K. by international standards. Interventions to reduce infection risk largely have been ignored by the DM community and should be a high priority for future research.”

ii. Poor Metabolic Control in Children and Adolescents With Type 1 Diabetes and Psychiatric Comorbidity. Some observations from the paper:

“Type 1 diabetes in childhood has been found to be associated with an increased risk of psychiatric comorbidities (13), which might intensify the burden of disease and accelerate metabolic deterioration (46), subsequently increasing the risk of mortality and long-term complications such as retinopathy, nephropathy, and neuropathy (79).

Metabolic dysregulation is closely linked to age and diabetes duration, showing a peak in adolescence and early adulthood (10,11). Early adolescence is also characterized as a time of psychological vulnerability (12), in which the incidence of major psychiatric disorders increases (13). A diagnosis of type 1 diabetes in early adolescence seems to increase psychological distress (1,2), and three large population-based studies have shown higher rates of psychiatric disorders in children and adolescents with type 1 diabetes compared with the general population (13). In particular, increased risk was seen for depression, anxiety, and eating disorders, where the pathogenesis is considered to involve reactive mechanisms and imbalances in the diathesis-stress system (13,14).”

“Despite clinical and research evidence that a child with type 1 diabetes often receives more than one psychiatric diagnosis (1,3), most studies evaluate one disorder at a time (46,1620). Motivated by findings that Danish children and adolescents with type 1 diabetes have a higher risk of developing a psychiatric disorder compared with the background population (2), we performed two studies based on the NPR and the Danish Registry of Childhood and Adolescent Diabetes (DanDiabKids). […] The NPR contains psychiatric and somatic diagnoses from all inpatient admissions to Danish public hospitals since 1977. […] The register has used the ICD-10 since 1994 (22,23). Data on registration of psychiatric and type 1 diabetes diagnoses were collected from the NPR, covering 1996 to April 2015. DanDiabKids collects information on all children and adolescents diagnosed with type 1 diabetes before the age of 15 years and monitors them until they are transferred to adult clinics at ∼18 years of age. All public hospital pediatric units must supply annual data on all patients with diabetes to DanDiabKids. […] DanDiabKids contains annual data on all registered patients since 1996, including information on quality indicators, demographic variables, associated conditions, diabetes classification, diabetes family history, growth, self-management, and treatment variables. DanDiabKids now covers 99% of all Danish children and adolescents diagnosed with type 1 diabetes before the age of 15 years. […] Our study population was generated by merging data from DanDiabKids and the NPR. The inclusion criteria were registration with type 1 diabetes in DanDiabKids, age at onset <15 years, year of onset 1995–2014, and year of birth after 1980.”

“After merging DanDiabKids with the NPR, 4,725 children and adolescents with type 1 diabetes were identified […]. Characteristics for the included subjects were as follows: mean age at onset of diabetes was 8.98 years (SD 3.81), birth year ranged from 1980 to 2013, mean age at last visit was 14.6 years (3.7), 2,462 (52.1%) were boys, mean duration of diabetes at last visit was 5.65 years (3.7), 4,434 (93.8%) were of Danish origin, 254 (5.4%) were immigrants or offspring of immigrants, and 36 (0.8%) had unknown ethnicity. […] The observed number of SH [severe hypoglycemia, US] and DKA events per 100 person-years was respectively 10.7 (SH) and 3.2 (DKA) in patients with neurodevelopmental/constitutional psychiatric disorder, 12.1 (SH) and 3.7 (DKA) in patients with potentially reactive psychiatric disorder, 12.3 (SH) and 6.4 (DKA) in patients with both types of psychiatric disorders, and 8.1 (SH) and 1.8 (DKA) in patients without psychiatric disorder. […] Among the 4,725 children and adolescents included in the study, 1,035 were diagnosed with at least one psychiatric disorder at some point. Of these, a total of 175 received their first psychiatric diagnosis before the onset of type 1 diabetes, 575 during pediatric care, and 285 were diagnosed after referral to adult care. […] Anxiety disorders were the most common (n = 492), followed by “behavioral and emotional disorders” (n = 310), mood disorders (n = 205), psychoactive substance misuse disorders (n = 190), and disorders of inattention and hyperactivity (ADHD/attention-deficit disorder [ADD]) (n = 172). Of the 1,035 patients, 46% were diagnosed with two or more psychiatric disorders and 22.8% were diagnosed with three or more psychiatric disorders.”

“Shortly after type 1 diabetes diagnosis, a higher estimated risk of psychiatric disorders was evident among patients who were 10–15 years old at onset of type 1 diabetes. However, after 15–20 years with diabetes, the differences among the groups leveled out at a risk of ∼30% […] Children with high mean HbA1c levels (>8.5% [>70 mmol/mol]) during the first 2 years showed the highest estimated risk of developing a psychiatric disorder, although these differences also appear to level out after 15–20 years with type 1 diabetes. […] The mean HbA1c level was higher in children with a psychiatric disorder (0.22% [95% CI 0.15; 0.29]; 2.45 mmol/mol [1.67; 3.22]) compared with children with no psychiatric disorder (P < 0.001) […] High HbA1c levels in the early period after type 1 diabetes onset seem to be a possible indicator for subsequent psychiatric disorders, and having a psychiatric disorder was associated with higher HbA1c levels, especially in patients with disorders of putative reactive pathogenesis. Given that the Kaplan-Meier plots showed that the estimated risk of being diagnosed with a psychiatric disorder within a period of 15–20 years of type 1 diabetes onset was close to 30% in most groups, our finding highlights an important clinical problem.”

“The estimated risk of developing a psychiatric disorder during the 15–20 years after type 1 diabetes diagnosis is high. The most vulnerable period appeared to be adolescence. Patients with poorly regulated diabetes shortly after onset had a higher estimated risk of developing psychiatric comorbidities. Young patients diagnosed with a psychiatric disorder had more episodes of DKA, and those diagnosed within the reactive spectrum had higher HbA1c levels. Children and adolescents with type 1 diabetes, and in particular those who fail to reach treatment goals, should be systematically evaluated regarding psychological vulnerabilities.”

iii. Development of Microvascular Complications and Effect of Concurrent Risk Factors in Type 1 Diabetes: A Multistate Model From an Observational Clinical Cohort Study.

“The prevalence of type 1 diabetes has increased over the past decades (1,2). Increased life expectancy means that people live longer with diabetes (35); thus, potentially more years are lived with both macrovascular and microvascular complications (6,7). Type 1 diabetes is a complex disease, which develops in various complication states, and co-occurrence of multiple microvascular complications frequently is seen (8). So far, most studies are of a single complication, and the association between the worsening of one complication and the incidence of another is well described, although independently of other complications (9,10). At the same time, a sizeable group of individuals seems to be protected from microvascular complications (1114), and some live several decades with type 1 diabetes without developing complications. Advanced statistical models, such as multistate models, offer an opportunity to explore the transition through various disease states and to quantify progression rates while considering the concurrent complication burden (15,16), that is, the complication burden at a given time point in the observation window.

Strong evidence indicates that some risk factors play a role in all types of microvascular complications. For example, the effects of the duration of diabetes and poor glycemic control are well documented (1720). For other risk factors, such as hypertension, an association has been established mainly for retinopathy and diabetic kidney disease (21,22). Adverse cholesterol levels and previous cardiovascular disease (CVD) are indisputably associated with a higher risk of macrovascular complications (23) and may play a role in the development of microvascular complications (24). […] The complex interplay between microvascular complications and risk factors has been explored only to a limited extent. In this study, we developed a multistate model of microvascular complications to describe in detail complication development in type 1 diabetes. We describe the development of sequences of diabetes-related microvascular complications at various states and examine the associations between selected risk factors, both alone and combined with existing complication burden, and incidence of (further) microvascular complications.”

“In total, 5,031 individuals with type 1 diabetes were registered at the SDCC during the study period. We excluded 1,203 because of missing data for diabetic kidney disease, retinopathy, and/or neuropathy, which left 3,828 eligible individuals to be included in the study. Of these, 242 were first seen in the final state with three complications, which left 3,586 available for analysis, corresponding to 22,946 person-years (PY) […] The median follow-up time was 7.8 years (25th–75th percentile 3.3–10.7 years). HbA1c level at the end of follow-up was lower than at entry, whereas the levels of blood pressure, lipids, and BMI were unchanged. An increase in the use of all cardioprotective medications was observed.”

“We identified 523 individuals who developed diabetic kidney disease during the study. Of these, 84 events occurred in individuals with no complications (IR 12.9 per 1,000 PY), 221 in individuals with retinopathy (25.7 per 1,000 PY), 27 in individuals with neuropathy (36.6 per 1,000 PY), and 191 in individuals with both neuropathy and retinopathy (61.8 per 1,000 PY). […] In the adjusted model, individuals with both retinopathy and neuropathy had a threefold higher risk of diabetic kidney disease than individuals without complications. […] A total of 482 individuals developed neuropathy during follow-up. Of these, 75 incidents occurred in individuals with no complications (IR 11.5 per 1,000 PY), 14 in individuals with diabetic kidney disease (20.6 per 1,000 PY), 234 in individuals with retinopathy (27.2 per 1,000 PY), and 159 in individuals with both retinopathy and diabetic kidney disease (50.2 per 1,000 PY). Individuals with both retinopathy and diabetic kidney disease had a 70% higher risk of developing neuropathy than individuals without complications […] In total, we recorded 649 individuals with incident retinopathy from any previous complication state. Of these, 459 incidents occurred in individuals with no complications (IR 70.7 per 1,000 PY), 74 in individuals with diabetic kidney disease (109.1 per 1,000 PY), 71 in individuals with neuropathy (96.6 per 1,000 PY), and 45 in individuals with both neuropathy and diabetic kidney disease (224.7 per 1,000 PY). Individuals with both diabetic kidney disease and neuropathy had a twofold higher IRR of developing retinopathy than individuals without complications”.

“Baseline and concurrent values of HbA1c, systolic blood pressure, eGFR, and baseline CVD status were all strongly associated with a higher risk of developing diabetic kidney disease. […] The analysis that included complication state revealed that individuals without any other complications than CVD had an almost three times higher risk of diabetic kidney disease than individuals without either CVD or microvascular complications. […] Duration of diabetes, baseline and concurrent value of HbA1c, systolic blood pressure, and baseline LDL cholesterol values were all factors associated with a higher risk of developing retinopathy. None of the effects of the modifiable risk factors on retinopathy were modified by complication burden. […] men with diabetic kidney disease had a higher risk of developing retinopathy than women with diabetic kidney disease. […] All investigated risk factors, except LDL cholesterol, were associated with incidence of neuropathy at both baseline and concurrent levels.”

“[W]e conducted a sensitivity analysis with retinopathy defined as severe nonproliferative or proliferative retinopathy. The prevalence and incidence of retinopathy were much lower, but all associations were similar to the main analysis […]. We found no effect modification by lipid-lowering or antihypertensive treatment. […] We found a stepwise higher risk of any microvascular complication in individuals with higher concurrent complication burden. Baseline and concurrent HbA1c levels, systolic blood pressure, and duration of diabetes were associated with the development of all three microvascular complications. For most risk factors, we did not find evidence that concurrent complication burden modified the association with complication development. […] Concurrent HbA1c level was a strong risk factor for all microvascular complications, even when we adjusted for age, duration, and other traditional risk factors. The overall effects were of similar magnitude to the effect of baseline levels of HbA1c and to other reports (11,29).”

“The presented results are interpreted in the frame of a multistate model design, and the use of clinical data makes the results highly relevant in similar health care settings. However, because of the observational study design, we cannot draw conclusions about causality. The positive associations among complications might reflect that diabetic kidney disease takes the longest time to develop, whereas retinopathy and neuropathy develop faster. Associations of two disease complications to a third might not be causal. However, that the risk of a third complication, even after adjustment for multiple confounders, is higher regardless of the previous combination of complications indicates that an association cannot be explained by these risk factors alone. In addition, concurrent risk factor levels may be subject to reverse causality. The current results should be seen as a benchmark for others who aim to explore the occurrence of microvascular complications as a function of the concurrent total complication burden in individuals with type 1 diabetes. […] The findings demonstrate that high concurrent complication burden elevates the risk of all three investigated microvascular complications: diabetic kidney disease, retinopathy, and neuropathy. This means that if an individual develops a complication, the clinician should be aware of the increased risk of developing more complications. […] For most risk factors, including HbA1c, we found no evidence that the effect on the development of microvascular complications was modified by the burden of concurrent complications.”

iv. Long-term Glycemic Control and Dementia Risk in Type 1 Diabetes.

“[P]rior work has established type 1 diabetes as a risk factor for dementia (15). However, the relationship between glycemic control and subsequent risk of dementia in those with type 1 diabetes remains unclear. Hemoglobin A1c (HbA1c) is an established measure that integrates glucose control over the prior 2–3 months and is widely used to guide clinical management of type 1 diabetes (16,17). Cumulative glycemic exposure, as measured by multiple HbA1c measures over time, has previously been used to evaluate glycemic trajectories and their association with a number of diabetes complications (18,19). Electronic health records capture HbA1c values collected over time allowing for a more thorough long-term characterization of glycemic exposure than is reflected by a single HbA1c measure. In this study, we leverage data [from northern California, US] collected over a span of 19 years to examine the association of cumulative glycemic exposure, as measured by repeated HbA1c values, with incident dementia among older adults with type 1 diabetes. We also examine the potential for a threshold of glycemic exposure above or below which risk of dementia increases.”

“The final analytic cohort consisted of 3,433 individuals (mean age at cohort entry = 56.1 years old; 47.1% female) […]. On average, individuals who developed dementia during follow-up were older at cohort entry (64.4 vs. 55.7 years) and were more likely to have a history of stroke (7.7% compared with 3.5%) at baseline. The mean follow-up time was 6.3 years (median 4.8 years [interquartile range (IQR) 1.7, 9.9]), and the mean number of HbA1c measurements was 13.5 (median 9.0 [IQR 3.0, 20.00]). By the end of follow-up on 30 September 2015, 155 members (4.5%) were diagnosed with dementia, 860 (25.1%) had a lapse of at least 90 days in membership coverage, 519 (15.1%) died without a dementia diagnosis, and 1,899 (55.3%) were still alive without dementia diagnosis. Among the 155 members who developed dementia over follow-up, the mean age at dementia diagnosis was 64.6 years (median 63.6 years [IQR 56.1, 72.3]).”

“In Cox proportional hazards models, dementia risk was higher in those with increased exposure to HbA1c 8–8.9% (64–74 mmol/mol) and ≥9% (≥75 mmol/mol) and lower in those with HbA1c 6–6.9% (42–52 mmol/mol) and 7–7.9% (53–63 mmol/mol). In fully adjusted models, compared with those with minimal exposure (<10% of HbA1c measurements) to HbA1c 8–8.9% and ≥9%, those with prolonged exposure (≥75% of measurements) were 2.51 and 2.13 times more likely to develop dementia, respectively (HbA1c 8–8.9% fully adjusted hazard ratio [aHR] 2.51 [95% CI 1.23, 5.11] and HbA1c ≥9% aHR 2.13 [95% CI 1.13, 4.01]) […]. In contrast, prolonged exposure to HbA1c 6–6.9 and 7–7.9% was associated with a 58% lower and 61% lower risk of dementia, respectively (HbA1c 6–6.9% aHR 0.42 [95% CI 0.21, 0.83] and HbA1c 7–7.9% aHR 0.39 [95% CI 0.18, 0.83]). […] Results were similar in Cox models examining cumulative glycemic exposure based on whether a majority (>50%) of an individual’s available HbA1c measurements fell into the following categories of HbA1c: <6, 6–6.9, 7–7.9, 8–8.9, and ≥9% […]. Majority exposure to HbA1c 8–8.9 and ≥9% was associated with an increased risk of dementia (HbA1c 8–8.9% aHR 1.65 [95% CI 1.06, 2.57] and HbA1c ≥9% aHR 1.79 [95% CI 1.11, 2.90]), while majority exposure to HbA1c 6–6.9 and 7–7.9% was associated with a reduced risk of dementia (HbA1c 6–6.9% aHR 0.55 [95% CI 0.34, 0.88] and HbA1c 7–7.9% aHR 0.55 [95% CI 0.37, 0.82]). Majority exposure to HbA1c <6% (<42 mmol/mol) was associated with increased dementia risk in age-adjusted models (HR 2.06 [95% CI 1.11, 3.82]), though findings did not remain significant in fully adjusted models (aHR 1.45 [95% CI 0.71, 2.92]). Findings were similar in sensitivity analyses among the subset of members who were ≥65 years of age at baseline (n = 1,082 [32% of the sample]), though the increased risk associated with majority time at HbA1c ≥9% was no longer statistically significant”.

“In this large sample of older adults with type 1 diabetes, we found that cumulative exposure to higher levels of HbA1c (8–8.9 and ≥9%) was associated with an increased risk of dementia, while cumulative exposure to well-controlled HbA1c (6–6.9 and 7–7.9%) was associated with a decreased risk of dementia. In fully adjusted models, compared with those with minimal exposure to HbA1c 8–8.9% and HbA1c ≥9%, those with prolonged exposure were more than twice as likely to develop dementia over the course of follow-up […]. By contrast, dementia risk was ∼60% lower among those with prolonged exposure to well-controlled HbA1c (6–6.9 and 7–7.9%) compared with those with minimal time at well-controlled levels of HbA1c.”

“Our results complement and extend previous studies that have reported an association between chronic hyperglycemia and decreased cognitive function in children and adolescents with type 1 diabetes (25,26), as well as studies reporting an association between poor glycemic control and decreased cognitive functioning in middle-aged adults with type 1 diabetes and older adults with type 2 diabetes (711). Our findings are also consistent with previous studies that found an increased dementia risk associated with poorer glycemic control among adults with type 2 diabetes and adults without diabetes (1113). Whether these findings applied to dementia risk among older adults with type 1 diabetes was previously unknown.”

“In our study of 3,433 older adults with type 1 diabetes, 155 (4.5%) individuals developed dementia over an average of 6.3 years of follow-up. Among those who developed dementia, the average age at dementia diagnosis was 64.6 years. A large-scale study using administrative health data from 1998 to 2011 in England reported a similar incidence of dementia among a subset of adults aged ≥50 years with type 1 diabetes (3.99% developed dementia), though the average length of follow-up was not reported for this specific age-group (15). Prior studies have also found type 1 diabetes to be a risk factor for dementia (15) and have reported the average age at onset of dementia to be 2–5 years earlier in those with diabetes compared with those without diabetes (27,28). Taken together, these results provide further evidence that older adults with type 1 diabetes are at increased risk of developing dementia and may have increased risk at younger ages than the general population. Our results, however, suggest that effective glycemic control could be an important tool for reducing risk of dementia among older adults with type 1 diabetes.”

“Pathophysiological mechanisms by which glycemic control may affect dementia risk are still poorly understood but are hypothesized to result from structural brain abnormalities stemming from chronic exposure to hyperglycemia and/or recurrent severe hypoglycemia. Studies in adults and youth with type 1 diabetes have reported an association between chronic hyperglycemia (defined using lifetime HbA1c history and using retinopathy as an indicator of chronic exposure) and gray matter density loss (3537). Studies examining the association between severe hypoglycemic events and changes in brain structure have been less consistent, with some reporting increased gray matter density loss and a higher prevalence of cortical atrophy in those with a history of frequent exposure to severe hypoglycemia (36,38), while another study reported no association (37). In the ACCORD MIND (Action to Control Cardiovascular Risk in Diabetes Memory in Diabetes) trial, compared with standard glycemic control, intensive glycemic control was associated with greater total brain volume, suggesting that intensive glycemic control may reduce brain atrophy related to diabetes (39). […] Understanding why glycemic patterns are associated with dementia is a much-needed area for future study, particularly with regard of the potential role of intercurrent micro- and macrovascular complications.”

v. A Comparison of the 2017 American College of Cardiology/American Heart Association Blood Pressure Guideline and the 2017 American Diabetes Association Diabetes and Hypertension Position Statement for U.S. Adults With Diabetes.

“Hypertension is one of the most common comorbidities among adults with diabetes. Prior studies have estimated the prevalence of hypertension to be twice as high among adults with diabetes compared with age-matched control subjects without diabetes (1,2). Among adults with diabetes, the presence of hypertension has been associated with a two times higher risk for cardiovascular disease (CVD) events and mortality (3,4).

The 2017 American College of Cardiology (ACC)/American Heart Association (AHA) Guideline for the Prevention, Detection, Evaluation, and Management of High Blood Pressure in Adults provides a comprehensive set of recommendations for the diagnosis and treatment of hypertension among adults, including those with diabetes (5). This guideline defines hypertension in adults, including those with diabetes, as an average systolic blood pressure (SBP) ≥130 mmHg or diastolic blood pressure (DBP) ≥80 mmHg […]. According to this guideline, pharmacological antihypertensive treatment should be initiated in adults with diabetes if they have an average SBP ≥130 mmHg or DBP ≥80 mmHg, and the treatment goal is SBP <130 mmHg and DBP <80 mmHg (5).

The American Diabetes Association (ADA) published a position statement on diabetes and hypertension in 2017 that recommends blood pressure (BP) levels different from the ACC/AHA guideline for defining hypertension and for initiating pharmacological antihypertensive treatment (for both, SBP ≥140 mmHg or DBP ≥90 mmHg) (6). The ADA position statement recommends that BP goals should be individualized based on patient priorities and clinician judgment. Treatment goals for those taking antihypertensive medication are SBP <140 mmHg and DBP <90 mmHg, with SBP <130 mmHg and DBP <80 mmHg to be considered for those with high CVD risk as long as these levels can be achieved without undo treatment burden.

The purpose of the current study was to estimate the impact of differences in the definition of hypertension and recommendations for pharmacological antihypertensive treatment initiation and intensification of therapy in U.S. adults with diabetes according to the ACC/AHA guideline and the ADA diabetes and hypertension position statement (5,6). To accomplish these goals, we analyzed data from the U.S. National Health and Nutrition Examination Survey (NHANES).”

“According to data from NHANES 2011–2016, 56.6% (95% CI 53.3, 59.9) of U.S. adults with diabetes were taking antihypertensive medication. Of U.S. adults with diabetes, 57.4% (53.1, 61.6) of those not taking and 80.2% (76.6, 83.4) of those taking antihypertensive medication had high CVD risk. Among U.S. adults with diabetes, those with high CVD risk (history of CVD or 10-year ASCVD risk ≥10%) were on average 15–20 years older and the prevalence of smoking and chronic kidney disease was 10–20% higher when compared with their counterparts without high CVD risk […]. Among U.S. adults with diabetes without high CVD risk, the mean 10-year and 30-year predicted CVD risks were 3.8% (3.5, 4.2) and 25.0% (23.4, 26.6), respectively, for those not taking antihypertensive medication and 5.8% (5.3, 6.4) and 37.4% (34.5, 40.3), respectively, for those taking antihypertensive medication.

The prevalence of hypertension was 77.1% (95% CI 73.9, 80.0) according to the ACC/AHA guideline and 66.3% (63.4, 69.1) according to the ADA position statement […]. Overall, 10.8% (9.0, 12.8) of U.S. adults with diabetes had hypertension according to the ACC/AHA guideline but not the ADA position statement. Among U.S. adults with diabetes not taking antihypertensive medication, 52.8% (47.7, 57.8), 24.8% (20.6, 29.6) and 22.4% (19.2, 25.9) were recommended antihypertensive medication initiation by neither document, by the 2017 ACC/AHA guideline only, and by both documents, respectively […]. Among U.S. adults with diabetes taking antihypertensive medication, 45.3% (41.3, 49.4), 4.3% (2.8, 6.6), and 50.4% (46.5, 54.2) had an average BP that met the goal in both documents, was above the ACC/AHA goal but not the ADA goal, and was above the goals in both documents, respectively […] The overall agreement between the ACC/AHA guideline and the ADA position statement was 89.2% (87.2, 91.0) for the presence of hypertension, 75.2% (70.4, 79.4) for the recommendation to initiate antihypertensive medication, and 95.7% (93.4, 97.2) for having a BP above the recommended treatment goal. “

“Based on both the ACC/AHA guideline and ADA position statement, 17.8 (95% CI 16.2, 19.3) million U.S. adults with diabetes had hypertension […]. An additional 2.9 (2.3, 3.5) million U.S. adults had hypertension based on the ACC/AHA guideline only. Among U.S. adults with diabetes not taking antihypertensive medication, 2.6 (2.1, 3.1) million were recommended to initiate antihypertensive medication by both the ACC/AHA guideline and the ADA position statement with an additional 2.9 (2.3, 3.5) million recommended to initiate antihypertensive medication by the ACC/AHA guideline only […]. Among U.S. adults with diabetes taking antihypertensive medication, 7.6 (6.8, 8.5) million had a BP above the goal in both documents, with an additional 700,000 (400,000, 900,000) having a BP above the goal recommended in the ACC/AHA guideline only […]. Among U.S. adults with diabetes not taking antihypertensive medication, the mean 10-year CVD risk was 10.7% (95% CI 9.4, 12.0) for those not recommended treatment initiation by either the ACC/AHA guideline or the ADA position statement, 14.6% (11.5, 17.6) for those recommended treatment initiation by the ACC/AHA guideline but not the ADA position statement, and 23.2% (19.5, 27.0) among those recommended treatment initiation by the ACC/AHA guideline and the ADA position statement […]. The mean 30-year CVD risk exceeded 25% in each of these groups. Among U.S. adults with diabetes taking antihypertensive medication, the mean 10-year CVD risk was 10.6% (9.4, 12.0), 6.5% (CI 5.6, 7.3), and 33.8% (32.1, 35.5) among those with above-goal BP according to neither document, the ACC/AHA guideline only, and both documents, respectively […]. The 30-year CVD risk exceeded 40% in each group.”

“In conclusion, the current study demonstrates a high degree of concordance between the 2017 ACC/AHA BP guideline and the 2017 ADA position statement on diabetes and hypertension. Using either document, the majority of U.S. adults with diabetes have hypertension. A substantial proportion of U.S. adults with diabetes not taking antihypertensive medication are recommended to initiate treatment by both documents […] Among U.S. adults with diabetes not taking antihypertensive medication, 75.2% had an identical recommendation for initiation of antihypertensive drug therapy according to the ACC/AHA guideline and the ADA position statement. The majority of those who were recommended to initiate pharmacological antihypertensive therapy according to the ACC/AHA guideline but not the ADA position statement had high CVD risk. […] At the population level, the ACC/AHA guideline and ADA position statement have more similarities than differences. However, at the individual level, some patients with diabetes will have fundamental changes in their care depending on which advice is followed. The decision to initiate and intensify antihypertensive medication should always be individualized, based on discussions between patients and their clinicians. Both the ACC/AHA BP guideline and ADA position statement acknowledge the need to individualize treatment decisions to align with patients’ interests.”

vi. Treatment-induced neuropathy of diabetes: an acute, iatrogenic complication of diabetes.

“Treatment-induced neuropathy in diabetes (also referred to as insulin neuritis) is considered a rare iatrogenic small fibre neuropathy caused by an abrupt improvement in glycaemic control in the setting of chronic hyperglycaemia. The prevalence and risk factors of this disorder are not known. In a retrospective review of all individuals referred to a tertiary care diabetic neuropathy clinic over 5 years, we define the proportion of individuals that present with and the risk factors for development of treatment-induced neuropathy in diabetes. Nine hundred and fifty-four individuals were evaluated for a possible diabetic neuropathy. Treatment-induced neuropathy in diabetes was defined as the acute onset of neuropathic pain and/or autonomic dysfunction within 8 weeks of a large improvement in glycaemic control—specified as a decrease in glycosylated haemoglobin A1C (HbA1c) of ≥2% points over 3 months. Detailed structured neurologic examinations, glucose control logs, pain scores, autonomic symptoms and other microvascular complications were measured every 3–6 months for the duration of follow-up. Of 954 patients evaluated for diabetic neuropathy, 104/954 subjects (10.9%) met criteria for treatment-induced neuropathy in diabetes with an acute increase in neuropathic or autonomic symptoms or signs coinciding with a substantial decrease in HbA1c. Individuals with a decrease in HbA1c had a much greater risk of developing a painful or autonomic neuropathy than those individuals with no change in HbA1c (P < 0.001), but also had a higher risk of developing retinopathy (P < 0.001) and microalbuminuria (P < 0.001). There was a strong correlation between the magnitude of decrease in HbA1c, the severity of neuropathic pain (R = 0.84, P < 0.001), the degree of parasympathetic dysfunction (R = −0.52, P < 0.01) and impairment of sympathetic adrenergic function as measured by fall in blood pressure on tilt-table testing (R = −0.63, P < 0.001). With a decrease in HbA1c of 2–3% points over 3 months there was a 20% absolute risk of developing treatment-induced neuropathy in diabetes, with a decrease in HbA1c of >4% points over 3 months the absolute risk of developing treatment-induced neuropathy in diabetes exceeded 80%. Treatment-induced neuropathy of diabetes is an underestimated iatrogenic disorder associated with diffuse microvascular complications. Rapid glycaemic change in patients with uncontrolled diabetes increases the risk of this complication.”

“Typically, individuals with TIND reported the onset of severe burning pain (pain scores 4–10/10) within 2–6 weeks of the improvement in glucose control. Burning pain was present in all subjects with TIND. Paraesthesias were present in 93/104 subjects and shooting pain in 88/104 subjects. Hyperalgesia and allodynia were common in the distribution of the pain. […] Individuals with TIND all reported ongoing sleep disturbances typically described as difficulty with sleep initiation and sleep duration secondary to neuropathic pain. These individuals reported no record of sleep problems prior to the development of TIND. […] Erectile dysfunction was noted in 28/31 males with TIND, compared to 135/417 males without TIND (P < 0.001, X2). […] Seventy-three individuals completed autonomic testing within 2–5 months of the onset of neuropathic pain. […] The results for both groups, in all tests, were abnormal compared to age-related normative values. There were strong correlations between the magnitude of decrease in HbA1c over 3 months and worsening autonomic function. A greater change in HbA1c resulted in worsening parasympathetic function as determined by the expiratory to inspiratory ratio (R = −0.52, P < 0.01) and the Valsalva ratio (R = −0.55, P < 0.01). Greater sympathetic adrenergic dysfunction also correlated with a greater change in HbA1c over 3 months as determined by the fall in systolic blood pressure during tilt-table test (R = −0.63, P < 0.001), the fall in blood pressure during phase 2 of the Valsalva manoeuvre (R = 0.49, P < 0.001), and the diminished phase 4 blood pressure overshoot during the Valsalva manoeuvre (R = −0.59, P < 0.001). […] individuals with type 1 diabetes had greater autonomic dysfunction than those with type 2 diabetes across all tests. The slopes of the regression lines describing the correlation between the change in HbA1c and a particular autonomic test did not differ by the type of diabetes, or by the type of treatment used to control glucose.”

“Most patients with TIND had rapid progression of retinopathy that developed in conjunction with the onset of neuropathic pain […] Prior to development of TIND, 65/104 individuals had no retinopathy, 35/104 had non-proliferative retinopathy, whereas 4/104 had proliferative retinopathy. Twelve months after the development of TIND, 10/104 individuals had no retinopathy, 54/104 had non-proliferative retinopathy and 40/104 had proliferative retinopathy (P < 0.001, Fisher’s exact test). Prior to development of TIND, 18/104 had evidence of microalbuminuria, while 12 months after the development of TIND, 87/104 had evidence of microalbuminuria (P < 0.001, X2).”

“TIND is a small fibre and autonomic neuropathy that appears after rapid improvements in glucose control. In this manuscript, we demonstrate that: (i) there is an unexpectedly high proportion of individuals with TIND in a tertiary referral diabetic clinic; (ii) the risk of developing TIND is associated with the magnitude and rate of change in HbA1c; (iii) neuropathic pain and autonomic dysfunction severity correlate with the magnitude of change in HbA1c; (iv) patients with Type 1 diabetes and a history of eating disorders are at high risk for developing TIND; and (v) TIND can occur with use of insulin or oral hypoglycaemic agents. […] TIND differs from the most prevalent generalized neuropathy of diabetes, the distal sensory-motor polyneuropathy, in several respects. The neuropathic pain has an acute onset, appearing within 8 weeks of glycaemic change, in contrast with the more insidious onset in the distal sensory-motor polyneuropathy […]. The pain in TIND is more severe, and poorly responsive to interventions including opioids, whereas most patients with distal sensory-motor polyneuropathy respond to non-opioid interventions […]. Although the distribution of the pain is length-dependent in individuals with TIND, it is frequently far more extensive than in distal sensory-motor polyneuropathy and the associated allodynia and hyperalgesia are much more prevalent […]. Autonomic symptoms and signs are common, prominent and appear acutely, in contrast to the relatively lower prevalence, gradual onset and slow progression in distal sensory-motor polyneuropathy […]. Finally, both the pain and autonomic features may be reversible in some patients […].

Our data indicate that the severity of TIND is associated with the magnitude of the change of HbA1c, however, it is also clear that the rate of change is important (e.g. a 4% point fall in the HbA1c will have a greater impact if occurring over 3 months than over 6 months). The pathogenic mechanisms whereby this change in glucose results in nerve damage and/or dysfunction are not known. Proposed mechanisms include endoneurial ischaemia due to epineurial arterio-venous shunts […], apoptosis due to glucose deprivation […], microvascular neuronal damage due to recurrent hypoglycaemia […], and ectopic firing of regenerating axon sprouts, but these possibilities are unproven. […] Additional mechanistic studies are necessary to determine the underlying pathophysiology.”

April 28, 2019 Posted by | Cardiology, Diabetes, Epidemiology, Immunology, Medicine, Nephrology, Neurology, Ophthalmology, Psychiatry, Studies | Leave a comment