Econstudentlog

A few diabetes papers of interest

i. Identical and Nonidentical Twins: Risk and Factors Involved in Development of Islet Autoimmunity and Type 1 Diabetes.

Some observations from the paper:

“Type 1 diabetes is preceded by the presence of preclinical, persistent islet autoantibodies (1). Autoantibodies against insulin (IAA) (2), GAD (GADA), insulinoma-associated antigen 2 (IA-2A) (3), and/or zinc transporter 8 (ZnT8A) (4) are typically present prior to development of symptomatic hyperglycemia and progression to clinical disease. These autoantibodies may develop many years before onset of type 1 diabetes, and increasing autoantibody number and titers have been associated with increased risk of progression to disease (57).

Identical twins have an increased risk of progression of islet autoimmunity and type 1 diabetes after one twin is diagnosed, although reported rates have been highly variable (30–70%) (811). This risk is increased if the proband twin develops diabetes at a young age (12). Concordance rates for type 1 diabetes in monozygotic twins with long-term follow-up is >50% (13). Risk for development of islet autoimmunity and type 1 diabetes for nonidentical twins is thought to be similar to non-twin siblings (risk of 6–10% for diabetes) (14). Full siblings who inherit both high-risk HLA (HLA DQA1*05:01 DR3/4*0302) haplotypes identical to their proband sibling with type 1 diabetes have a much higher risk for development of diabetes than those who share only one or zero haplotypes (55% vs. 5% by 12 years of age, respectively; P = 0.03) (15). Despite sharing both HLA haplotypes with their proband, siblings without the HLA DQA1*05:01 DR3/4*0302 genotype had only a 25% risk for type 1 diabetes by 12 years of age (15).”

“The TrialNet Pathway to Prevention Study (previously the TrialNet Natural History Study; 16) has been screening relatives of patients with type 1 diabetes since 2004 and follows these subjects with serial autoantibody testing for the development of islet autoantibodies and type 1 diabetes. The study offers longitudinal monitoring for autoantibody-positive subjects through HbA1c testing and oral glucose tolerance tests (OGTTs).”

“The purpose of this study was to evaluate the prevalence of islet autoantibodies and analyze a logistic regression model to test the effects of genetic factors and common twin environment on the presence or absence of islet autoantibodies in identical twins, nonidentical twins, and full siblings screened in the TrialNet Pathway to Prevention Study. In addition, this study analyzed the presence of islet autoantibodies (GADA, IA-2A, and IAA) and risk of type 1 diabetes over time in identical twins, nonidentical twins, and full siblings followed in the TrialNet Pathway to Prevention Study. […] A total of 48,051 sibling subjects were initially screened (288 identical twins, 630 nonidentical twins, and 47,133 full siblings). Of these, 48,026 had an initial screening visit with GADA, IA2A, and IAA results (287 identical twins, 630 nonidentical twins, and 47,109 full siblings). A total of 17,226 participants (157 identical twins, 283 nonidentical twins and 16,786 full siblings) were followed for a median of 2.1 years (25th percentile 1.1 year and 75th percentile 4.0 years), with follow-up defined as at least ≥12 months follow-up after initial screening visit.”

“At the initial screening visit, GADA was present in 20.2% of identical twins (58 out of 287), 5.6% of nonidentical twins (35 out of 630), and 4.7% of full siblings (2,205 out of 47,109) (P < 0.0001). Additionally, IA-2A was present primarily in identical twins (9.4%; 27 out of 287) and less so in nonidentical twins (3.3%; 21 out of 630) and full siblings (2.2%; 1,042 out of 47,109) (P = 0.0001). Nearly 12% of identical twins (34 out of 287) were positive for IAA at initial screen, whereas 4.6% of nonidentical twins (29 out of 630) and 2.5% of full siblings (1,152 out of 47,109) were initially IAA positive (P < 0.0001).”

“At 3 years of follow-up, the risk for development of GADA was 16% for identical twins, 5% for nonidentical twins, and 4% for full siblings (P < 0.0001) (Fig. 1A). The risk for development of IA-2A by 3 years of follow-up was 7% for identical twins, 4% for nonidentical twins, and 2% for full siblings (P = 0.0005) (Fig. 1B). At 3 years of follow-up, the risk of development of IAA was 10% for identical twins, 5% for nonidentical twins, and 4% for full siblings (P = 0.006) […] In initially autoantibody-negative subjects, 1.5% of identical twins, 0% of nonidentical twins, and 0.5% of full siblings progressed to diabetes at 3 years of follow-up (P = 0.18) […] For initially single autoantibody–positive subjects, at 3 years of follow-up, 69% of identical twins, 13% of nonidentical twins, and 12% of full siblings developed type 1 diabetes (P < 0.0001) […] Subjects who were positive for multiple autoantibodies at screening had a higher risk of developing type 1 diabetes at 3 years of follow-up with 69% of identical twins, 72% of nonidentical twins, and 47% of full siblings developing type 1 diabetes (P = 0.079)”

“Because TrialNet is not a birth cohort and the median age at screening visit was 11 years overall, this study would not capture subjects who had initial seroconversion at a young age and then progressed through the intermediate stage of multiple antibody positivity before developing diabetes.”

“This study of >48,000 siblings of patients with type 1 diabetes shows that at initial screening, identical twins were more likely to have at least one positive autoantibody and be positive for GADA, IA-2A, and IAA than either nonidentical twins or full siblings. […] risk for development of type 1 diabetes at 3 years of follow-up was high for both single and multiple autoantibody–positive identical twins (62–69%) and multiple autoantibody–positive nonidentical twins (72%) compared with 47% for initially multiple autoantibody–positive full siblings and 12–13% for initially single autoantibody–positive nonidentical twins and full siblings. To our knowledge, this is the largest prediagnosis study to evaluate the effects of genetic factors and common twin environment on the presence or absence of islet autoantibodies.

In this study, younger age, male sex, and genetic factors were significantly associated with expression of IA-2A, IAA, more than one autoantibody, and more than two autoantibodies, whereas only genetic factors were significant for GADA. An influence of common twin environment (E) was not seen. […] Previous studies have shown that identical twin siblings of patients with type 1 diabetes have a higher concordance rate for development of type 1 diabetes compared with nonidentical twins, although reported rates for identical twins have been highly variable (30–70%) […]. Studies from various countries (Australia, Denmark, Finland, Great Britain, and U.S.) have reported concordance rates for nonidentical twins ∼5–15% […]. Concordance rates have been higher when the proband was diagnosed at a younger age (8), which may explain the variability in these reported rates. In this study, autoantibody-negative nonidentical and identical twins had a low risk of type 1 diabetes by 3 years of follow-up. In contrast, once twins developed autoantibodies, risk for type 1 diabetes was high for multiple autoantibody nonidentical twins and both single and multiple autoantibody identical twins.”

ii. A Type 1 Diabetes Genetic Risk Score Can Identify Patients With GAD65 Autoantibody–Positive Type 2 Diabetes Who Rapidly Progress to Insulin Therapy.

This is another paper in the ‘‘ segment from the February edition of Diabetes Care – multiple other papers on related topics were also included in that edition, so if you’re interested in the genetics of diabetes it may be worth checking out.

Some observations from the paper:

“Type 2 diabetes is a progressive disease due to a gradual reduction in the capacity of the pancreatic islet cells (β-cells) to produce insulin (1). The clinical course of this progression is highly variable, with some patients progressing very rapidly to requiring insulin treatment, whereas others can be successfully treated with lifestyle changes or oral agents for many years (1,2). Being able to identify patients likely to rapidly progress may have clinical utility in prioritization monitoring and treatment escalation and in choice of therapy.

It has previously been shown that many patients with clinical features of type 2 diabetes have positive GAD65 autoantibodies (GADA) and that the presence of this autoantibody is associated with faster progression to insulin (3,4). This is often termed latent autoimmune diabetes in adults (LADA) (5,6). However, the predictive value of GADA testing is limited in a population with clinical type 2 diabetes, with many GADA-positive patients not requiring insulin treatment for many years (4,7). Previous research has suggested that genetic variants in the HLA region associated with type 1 diabetes are associated with more rapid progression to insulin in patients with clinically defined type 2 diabetes and positive GADA (8).

We have recently developed a type 1 diabetes genetic risk score (T1D GRS), which provides an inexpensive ($70 in our local clinical laboratory and <$20 where DNA has been previously extracted), integrated assessment of a person’s genetic susceptibility to type 1 diabetes (9). The score is composed of 30 type 1 diabetes risk variants weighted for effect size and aids discrimination of type 1 diabetes from type 2 diabetes. […] We aimed to determine if the T1D GRS could predict rapid progression to insulin (within 5 years of diagnosis) over and above GADA testing in patients with a clinical diagnosis of type 2 diabetes treated without insulin at diagnosis.”

“We examined the relationship between GADA, T1D GRS, and progression to insulin therapy using survival analysis in 8,608 participants with clinical type 2 diabetes initially treated without insulin therapy. […] In this large study of participants with a clinical diagnosis of type 2 diabetes, we have found that type 1 genetic susceptibility alters the clinical implications of a positive GADA when predicting rapid time to insulin. GADA-positive participants with high T1D GRS were more likely to require insulin within 5 years of diagnosis, with 48% progressing to insulin in this time in contrast to only 18% in participants with low T1D GRS. The T1D GRS was independent of and additive to participant’s age of diagnosis and BMI. However, T1D GRS was not associated with rapid insulin requirement in participants who were GADA negative.”

“Our findings have clear implications for clinical practice. The T1D GRS represents a novel clinical test that can be used to enhance the prognostic value of GADA testing. For predicting future insulin requirement in patients with apparent type 2 diabetes who are GADA positive, T1D GRS may be clinically useful and can be used as an additional test in the screening process. However, in patients with type 2 diabetes who are GADA negative, there is no benefit gained from genetic testing. This is unsurprising, as the prevalence of underlying autoimmunity in patients with a clinical phenotype of type 2 diabetes who are GADA negative is likely to be extremely low; therefore, most GADA-negative participants with high T1D GRS will have nonautoimmune diabetes. The use of this two-step testing approach may facilitate a precision medicine approach to patients with apparent type 2 diabetes; patients who are likely to progress rapidly are identified for targeted management, which may include increased monitoring, early therapy intensification, and/or interventions aimed at slowing progression (36,37).

The costs of analyzing the T1D GRS are relatively modest and may fall further, as genetic testing is rapidly becoming less expensive (38). […] In conclusion, a T1D GRS alters the clinical implications of a positive GADA test in patients with clinical type 2 diabetes and is independent of and additive to clinical features. This therefore represents a novel test for identifying patients with rapid progression in this population.”

iii. Retinopathy and RAAS Activation: Results From the Canadian Study of Longevity in Type 1 Diabetes.

“Diabetic retinopathy is the most common cause of preventable blindness in individuals ages 20–74 years and is the most common vascular complication in type 1 and type 2 diabetes (13). On the basis of increasing severity, diabetic retinopathy is classified into nonproliferative diabetic retinopathy (NPDR), defined in early stages by the presence of microaneurysms, retinal vascular closure, and alteration, or proliferative diabetic retinopathy (PDR), defined by the growth of new aberrant blood vessels (neovascularization) susceptible to hemorrhage, leakage, and fibrosis (4). Diabetic macular edema (DME) can be present at any stage of retinopathy and is characterized by increased vascular permeability leading to retinal thickening.

Important risk factors for the development of retinopathy continue to be chronic hyperglycemia, hyperlipidemia, hypertension, and diabetes duration (5,6). Given the systemic nature of these risk factors, cooccurrence of retinopathy with other vascular complications is common in patients with diabetes.”

“A key pathway implicated in diabetes-related small-vessel disease is overactivation of neurohormones. Activation of the neurohormonal renin-angiotensin-aldosterone system (RAAS) pathway predominates in diabetes in response to hyperglycemia and sodium retention. The RAAS plays a pivotal role in regulating systemic BP through vasoconstriction and fluid-electrolyte homeostasis. At the tissue level, angiotensin II (ANGII), the principal mediator of the RAAS, is implicated in fibrosis, oxidative stress, endothelial damage, thrombosis, inflammation, and vascular remodeling. Of note, systemic RAAS blockers reduce the risk of progression of eye disease but not DKD [Diabetic Kidney Disease, US] in adults with type 1 diabetes with normoalbuminuria (12).

Several longitudinal epidemiologic studies of diabetic retinopathy have been completed in type 1 diabetes; however, few have studied the relationships between eye, nerve, and renal complications and the influence of RAAS activation after prolonged duration (≥50 years) in adults with type 1 diabetes. As a result, less is known about mechanisms that persist in diabetes-related microvascular complications after long-standing diabetes. Accordingly, in this cross-sectional analysis from the Canadian Study of Longevity in Type 1 Diabetes involving adults with type 1 diabetes for ≥50 years, our aims were to phenotype retinopathy stage and determine associations between the presence of retinopathy and other vascular complications. In addition, we examined the relationship between retinopathy stage and renal and systemic hemodynamic function, including arterial stiffness, at baseline and dynamically after RAAS activation with an infusion of exogenous ANGII.”

“Of the 75 participants, 12 (16%) had NDR [no diabetic retinopathy], 24 (32%) had NPDR, and 39 (52%) had PDR […]. At baseline, those with NDR had lower mean HbA1c compared with those with NPDR and PDR (7.4 ± 0.7% and 7.5 ± 0.9%, respectively; P for trend = 0.019). Of note, those with more severe eye disease (PDR) had lower systolic and diastolic BP values but a significantly higher urine albumin-to-creatine ratio (UACR) […] compared with those with less severe eye disease (NPDR) or with NDR despite higher use of RAAS inhibitors among those with PDR compared with NPDR or NDR. History of cardiovascular and peripheral vascular disease history was significantly higher in participants with PDR (33.3%) than in those with NPDR (8.3%) or NDR (0%). Diabetic sensory polyneuropathy was prevalent across all groups irrespective of retinopathy status but was numerically higher in the PDR group (95%) than in the NPDR (86%) or NDR (75%) groups. No significant differences were observed in retinal thickness across the three groups.”

One quick note: This was mainly an eye study, but some of the other figures here are well worth taking note of. 3 out of 4 people in the supposedly low-risk group without eye complications had sensory polyneuropathy after 50 years of diabetes.

Conclusions

Hyperglycemia contributes to the pathogenesis of diabetic retinopathy through multiple interactive pathways, including increased production of advanced glycation end products, IGF-I, vascular endothelial growth factor, endothelin, nitric oxide, oxidative damage, and proinflammatory cytokines (2933). Overactivation of the RAAS in response to hyperglycemia also is implicated in the pathogenesis of diabetes-related complications in the retina, nerves, and kidney and is an important therapeutic target in type 1 diabetes. Despite what is known about these underlying pathogenic mechanisms in the early development of diabetes-related complications, whether the same mechanisms are active in the setting of long-standing type 1 diabetes is not known. […] In this study, we observed that participants with PDR were more likely to be taking RAAS inhibitors, to have a higher frequency of cardiovascular or peripheral vascular disease, and to have higher UACR levels, likely reflecting the higher overall risk profile of this group. Although it is not possible to determine why some patients in this cohort developed PDR while others did not after similar durations of type 1 diabetes, it seems unlikely that glycemic control alone is sufficient to fully explain the observed between-group differences and differing vascular risk profiles. Whereas the NDR group had significantly lower mean HbA1c levels than the NPDR and PDR groups, differences between participants with NPDR and those with PDR were modest. Accordingly, other factors, such as differences in vascular function, neurohormones, growth factors, genetics, and lifestyle, may play a role in determining retinopathy severity at the individual level.

The association between retinopathy and risk for DKD is well established in diabetes (34). In the setting of type 2 diabetes, patients with high levels of UACR have twice the risk of developing diabetic retinopathy than those with normal UACR levels. For example, Rodríguez-Poncelas et al. (35) demonstrated that impaired renal function is linked with increased diabetic retinopathy risk. Consistent with these studies and others, the PDR group in this Canadian Study of Longevity in Type 1 Diabetes demonstrated significantly higher UACR, which is associated with an increased risk of DKD progression, illustrating that the interaction between eye and kidney disease progression also may exist in patients with long-standing type 1 diabetes. […] In conclusion, retinopathy was prevalent after prolonged type 1 diabetes duration, and retinopathy severity associated with several measures of neuropathy and with higher UACR. Differential exaggerated responses to RAAS activation in the peripheral vasculature of the PDR group highlights that even in the absence of DKD, neurohormonal abnormalities are likely still operant, and perhaps accentuated, in patients with PDR even after long-standing type 1 diabetes duration.”

iv. Clinical and MRI Features of Cerebral Small-Vessel Disease in Type 1 Diabetes.

“Type 1 diabetes is associated with a fivefold increased risk of stroke (1), with cerebral small-vessel disease (SVD) as the most common etiology (2). Cerebral SVD in type 1 diabetes, however, remains scarcely investigated and is challenging to study in vivo per se owing to the size of affected vasculature (3); instead, MRI signs of SVD are studied. In this study, we aimed to assess the prevalence of cerebral SVD in subjects with type 1 diabetes compared with healthy control subjects and to characterize diabetes-related variables associated with SVD in stroke-free people with type 1 diabetes.”

RESEARCH DESIGN AND METHODS This substudy was cross-sectional in design and included 191 participants with type 1 diabetes and median age 40.0 years (interquartile range 33.0–45.1) and 30 healthy age- and sex-matched control subjects. All participants underwent clinical investigation and brain MRIs, assessed for cerebral SVD.

RESULTS Cerebral SVD was more common in participants with type 1 diabetes than in healthy control subjects: any marker 35% vs. 10% (P = 0.005), cerebral microbleeds (CMBs) 24% vs. 3.3% (P = 0.008), white matter hyperintensities 17% vs. 6.7% (P = 0.182), and lacunes 2.1% vs. 0% (P = 1.000). Presence of CMBs was independently associated with systolic blood pressure (odds ratio 1.03 [95% CI 1.00–1.05], P = 0.035).”

Conclusions

Cerebral SVD is more common in participants with type 1 diabetes than in healthy control subjects. CMBs especially are more prevalent and are independently associated with hypertension. Our results indicate that cerebral SVD starts early in type 1 diabetes but is not explained solely by diabetes-related vascular risk factors or the generalized microvascular disease that takes place in diabetes (7).

There are only small-scale studies on cerebral SVD, especially CMBs, in type 1 diabetes. Compared with the current study, one study with similar diabetes characteristics (i.e., diabetes duration, glycemic control, and blood pressure levels) as in the current study, but lacking a control population, showed a higher prevalence of WMHs, with more than half of the participants affected, but similar prevalence of lacunes and lower prevalence of CMBs (8). In another study, including 67 participants with type 1 diabetes and 33 control subjects, there was no difference in WMH prevalence but a higher prevalence of CMBs in participants with type 1 diabetes and retinopathy compared with control subjects (9). […] In type 1 diabetes, albuminuria and systolic blood pressure independently increase the risk for both ischemic and hemorrhagic stroke (12). […] We conclude that cerebral SVD is more common in subjects with type 1 diabetes than in healthy control subjects. Future studies will focus on longitudinal development of SVD in type 1 diabetes and the associations with brain health and cognition.”

v. The Legacy Effect in Type 2 Diabetes: Impact of Early Glycemic Control on Future Complications (The Diabetes & Aging Study).

“In the U.S., an estimated 1.4 million adults are newly diagnosed with diabetes every year and present an important intervention opportunity for health care systems. In patients newly diagnosed with type 2 diabetes, the benefits of maintaining an HbA1c <7.0% (<53 mmol/mol) are well established. The UK Prospective Diabetes Study (UKPDS) found that a mean HbA1c of 7.0% (53 mmol/mol) lowers the risk of diabetes-related end points by 12–32% compared with a mean HbA1c of 7.9% (63 mmol/mol) (1,2). Long-term observational follow-up of this trial revealed that this early glycemic control has durable effects: Reductions in microvascular events persisted, reductions in cardiovascular events and mortality were observed 10 years after the trial ended, and HbA1c values converged (1). Similar findings were observed in the Diabetes Control and Complications Trial (DCCT) in patients with type 1 diabetes (24). These posttrial observations have been called legacy effects (also metabolic memory) (5), and they suggest the importance of early glycemic control for the prevention of future complications of diabetes. Although these clinical trial long-term follow-up studies demonstrated legacy effects, whether legacy effects exist in real-world populations, how soon after diabetes diagnosis legacy effects may begin, or for what level of glycemic control legacy effects may exist are not known.

In a previous retrospective cohort study, we found that patients with newly diagnosed diabetes and an initial 10-year HbA1c trajectory that was unstable (i.e., changed substantially over time) had an increased risk for future microvascular events, even after adjusting for HbA1c exposure (6). In the same cohort population, this study evaluates associations between the duration and intensity of glycemic control immediately after diagnosis and the long-term incidence of future diabetic complications and mortality. We hypothesized that a glycemic legacy effect exists in real-world populations, begins as early as the 1st year after diabetes diagnosis, and depends on the level of glycemic exposure.”

RESEARCH DESIGN AND METHODS This cohort study of managed care patients with newly diagnosed type 2 diabetes and 10 years of survival (1997–2013, average follow-up 13.0 years, N = 34,737) examined associations between HbA1c <6.5% (<48 mmol/mol), 6.5% to <7.0% (48 to <53 mmol/mol), 7.0% to <8.0% (53 to <64 mmol/mol), 8.0% to <9.0% (64 to <75 mmol/mol), or ≥9.0% (≥75 mmol/mol) for various periods of early exposure (0–1, 0–2, 0–3, 0–4, 0–5, 0–6, and 0–7 years) and incident future microvascular (end-stage renal disease, advanced eye disease, amputation) and macrovascular (stroke, heart disease/failure, vascular disease) events and death, adjusting for demographics, risk factors, comorbidities, and later HbA1c.

RESULTS Compared with HbA1c <6.5% (<48 mmol/mol) for the 0-to-1-year early exposure period, HbA1c levels ≥6.5% (≥48 mmol/mol) were associated with increased microvascular and macrovascular events (e.g., HbA1c 6.5% to <7.0% [48 to <53 mmol/mol] microvascular: hazard ratio 1.204 [95% CI 1.063–1.365]), and HbA1c levels ≥7.0% (≥53 mmol/mol) were associated with increased mortality (e.g., HbA1c 7.0% to <8.0% [53 to <64 mmol/mol]: 1.290 [1.104–1.507]). Longer periods of exposure to HbA1c levels ≥8.0% (≥64 mmol/mol) were associated with increasing microvascular event and mortality risk.

CONCLUSIONS Among patients with newly diagnosed diabetes and 10 years of survival, HbA1c levels ≥6.5% (≥48 mmol/mol) for the 1st year after diagnosis were associated with worse outcomes. Immediate, intensive treatment for newly diagnosed patients may be necessary to avoid irremediable long-term risk for diabetic complications and mortality.”

Do note that the effect sizes here are very large and this stuff seems really quite important. Judging from the results of this study, if you’re newly diagnosed and you only obtain a HbA1c of say, 7.3% in the first year, that may translate into a close to 30% increased risk of death more than 10 years into the future, compared to a scenario of an HbA1c of 6.3%. People who did not get their HbA1c measured within the first 3 months after diagnosis had a more than 20% increased risk of mortality during the study period. This seems like critical stuff to get right.

vi. Event Rates and Risk Factors for the Development of Diabetic Ketoacidosis in Adult Patients With Type 1 Diabetes: Analysis From the DPV Registry Based on 46,966 Patients.

“Diabetic ketoacidosis (DKA) is a life-threatening complication of type 1 diabetes mellitus (T1DM) that results from absolute insulin deficiency and is marked by acidosis, ketosis, and hyperglycemia (1). Therefore, prevention of DKA is one goal in T1DM care, but recent data indicate increased incidence (2).

For adult patients, only limited data are available on rates and risk factors for development of DKA, and this complication remains epidemiologically poorly characterized. The Diabetes Prospective Follow-up Registry (DPV) has followed patients with diabetes from 1995. Data for this study were collected from 2000 to 2016. Inclusion criteria were diagnosis of T1DM, age at diabetes onset ≥6 months, patient age at follow-up ≥18 years, and diabetes duration ≥1 year to exclude DKA at manifestation. […] In total, 46,966 patients were included in this study (average age 38.5 years [median 21.2], 47.6% female). The median HbA1c was 7.7% (61 mmol/mol), median diabetes duration was 13.6 years, and 58.3% of the patients were treated in large diabetes centers.

On average, 2.5 DKA-related hospital admissions per 100 patient-years (PY) were observed (95% CI 2.1–3.0). The rate was highest in patients aged 18–30 years (4.03/100 PY) and gradually declined with increasing age […] No significant differences between males (2.46/100 PY) and females (2.59/100 PY) were found […] Patients with HbA1c levels <7% (53 mmol/mol) had significantly fewer DKA admissions than patients with HbA1c ≥9% (75 mmol/mol) (0.88/100 PY vs. 6.04/100 PY; P < 0.001)”

“Regarding therapy, use of an insulin pump (continuous subcutaneous insulin infusion [CSII]) was not associated with higher DKA rates […], while patients aged 31–50 years on CSII showed lower rates than patients using multiple daily injections (2.21 vs. 3.12/100 PY; adjusted P < 0.05) […]. Treatment in a large center was associated with lower DKA-related hospital admissions […] In both adults and children, poor metabolic control was the strongest predictor of hospital admission due to DKA. […] In conclusion, the results of this study identify patients with T1DM at risk for DKA (high HbA1c, diabetes duration 5–10 years, migrants, age 30 years and younger) in real-life diabetes care. These at-risk individuals may need specific attention since structured diabetes education has been demonstrated to specifically reduce and prevent this acute complication.”

August 13, 2019 - Posted by | Cardiology, Diabetes, Genetics, Immunology, Medicine, Molecular biology, Nephrology, Neurology, Ophthalmology, Studies

No comments yet.

Leave a comment

This site uses Akismet to reduce spam. Learn how your comment data is processed.