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 | Leave a comment

A few diabetes papers of interest

i. The dynamic origins of type 1 diabetes.

“Over a century ago, there was diabetes and only diabetes. Subsequently, diabetes came to be much more discretely defined (1) by age at onset (childhood or adult onset), clinical phenotype (lean or obese), treatment (insulin dependent or not insulin dependent), and, more recently, immune genotype (type 1 or type 2 diabetes). Although these categories broadly describe groups, they are often insufficient to categorize specific individuals, such as children having non–insulin-dependent diabetes and adults having type 1 diabetes (T1D) even when not requiring insulin. Indeed, ketoacidosis at presentation can be a feature of either T1D or type 2 diabetes. That heterogeneity extends to the origins and character of both major types of diabetes. In this issue of Diabetes Care, Redondo et al. (2) leverage the TrialNet study of subjects with a single diabetes-associated autoantibody at screening in order to explore factors determining progression to multiple autoantibodies and, subsequently, the pathogenesis of T1D.

T1D is initiated by presumed nongenetic event(s) operating in children with potent genetic susceptibility. But there is substantial heterogeneity even within the origins of this disease. Those nongenetic events evoke different autoantibodies such that T1D patients with insulin autoantibodies (IAA) have different features from those with GAD autoantibodies (GADA) (3,4). The former, in contrast with the latter, are younger both at seroconversion and at development of clinical diabetes, the two groups having different genetic risk and those with IAA having greater insulin secretory loss […]. These observations hint at distinct disease-associated networks leading to T1D, perhaps induced by distinct nongenetic events. Such disease-associated pathways could operate in unison, especially in children with T1D, who often have multiple autoantibodies. […]

Genetic analyses of autoimmune diseases suggest that only a small number of pathways contribute to disease risk. These pathways include NF-κB signaling, T-cell costimulation, interleukin-2, and interleukin-21 pathways and type 1 interferon antiviral responses (5,6). T1D shares most risk loci with celiac disease and rheumatoid arthritis (5), while paradoxically most risk loci shared with inflammatory bowel disease are protective or involve different haplotypes at the same locus. […] Events leading to islet autoimmunity may be encountered very early in life and invoke disease risk or disease protection (4,7) […]. Islet autoantibodies rarely appear before age 6 months, and among children with a family history of T1D there are two peaks for autoantibody seroconversion (3,4), the first for IAA at approximately age 1–2 years, while GADA-restricted autoimmunity develops after age 3 years up to adolescence, with a peak at about age 11 years”

“The precise nature of […] disease-associated nongenetic events remains unclear, but knowledge of the disease heterogeneity (1,9) has cast light on their character. Nongenetic events are implicated in increasing disease incidence, disease discordance even between identical twins, and geographical variation; e.g., Finland has 100-fold greater childhood T1D incidence than China (9,10). That effect likely increases with older age at onset […] disease incidence in Finland is sixfold greater than in an adjacent, relatively impoverished Russian province, despite similar racial origins and frequencies of high-risk HLA DQ genotypes […] Viruses, especially enteroviruses, and dietary factors have been invoked (1215). The former have been implicated because of the genetic association with antiviral interferon networks, seasonal pattern of autoantibody conversion, seroconversion being associated with enterovirus infections, and protection from seroconversion by maternal gestational respiratory infection, while respiratory infections even in the first year of life predispose to seroconversion (14) […]. Dietary factors also predispose to seroconversion and include the time of introduction of solid foods and the use of vitamin C and vitamin D (13,15). The Diabetes Autoimmunity Study in the Young (DAISY) found that early exposure to solid food (1–3 months of age) and vitamin C and late exposure to vitamin D and gluten (after 6 and 9 months of age, respectively) are T1D risk factors, leading the researchers to suggest that genetically at-risk children should have solid foods introduced at about 4 months of age with a diet high in dairy and fruit (13).” [my bold, US]

“This TCF7L2 locus is of particular interest in the context of T1D (9) as it is usually seen as the major type 2 diabetes signal worldwide. The rs7903146 SNP optimally captures that TCF7L2 disease association and is likely the causal variant. Intriguingly, this locus is associated, in some populations, with those adult-onset autoimmune diabetes patients with GADA alone who masquerade as having type 2 diabetes, since they initially do not require insulin therapy, and also markedly increases the diabetes risk in cystic fibrosis patients. One obvious explanation for these associations is that adult-onset autoimmune diabetes is simply a heterogeneous disease, an admixture of both T1D and type 2 diabetes (9), in which shared genes alter the threshold for diabetes. […] A high proportion of T1D cases present in adulthood (17,18), likely more than 50%, and many do not require insulin initially. The natural history, phenotype, and metabolic changes in adult-onset diabetes with GADA resemble a separate cluster of cases with type 2 diabetes but without GADA, which together constitute up to 24% of adult-onset diabetes (19). […] Knowledge of heterogeneity enables understanding of disease processes. In particular, identification of distinct pathways to clinical diabetes offers the possibility of defining distinct nongenetic events leading to T1D and, by implication, modulating those events could limit or eliminate disease progression. There is a growing appreciation that the two major types of diabetes may share common etiopathological factors. Just as there are a limited number of genes and pathways contributing to autoimmunity risk, there may also be a restricted number of pathways contributing to β-cell fragility.”

ii. The Association of Severe Diabetic Retinopathy With Cardiovascular Outcomes in Long-standing Type 1 Diabetes: A Longitudinal Follow-up.

OBJECTIVE It is well established that diabetic nephropathy increases the risk of cardiovascular disease (CVD), but how severe diabetic retinopathy (SDR) impacts this risk has yet to be determined.

RESEARCH DESIGN AND METHODS The cumulative incidence of various CVD events, including coronary heart disease (CHD), peripheral artery disease (PAD), and stroke, retrieved from registries, was evaluated in 1,683 individuals with at least a 30-year duration of type 1 diabetes drawn from the Finnish Diabetic Nephropathy Study (FinnDiane).”

RESULTS During 12,872 person-years of follow-up, 416 incident CVD events occurred. Even in the absence of DKD [Diabetic Kidney Disease], SDR increased the risk of any CVD (hazard ratio 1.46 [95% CI 1.11–1.92]; P < 0.01), after adjustment for diabetes duration, age at diabetes onset, sex, smoking, blood pressure, waist-to-hip ratio, history of hypoglycemia, and serum lipids. In particular, SDR alone was associated with the risk of PAD (1.90 [1.13–3.17]; P < 0.05) and CHD (1.50 [1.09–2.07; P < 0.05) but not with any stroke. Moreover, DKD increased the CVD risk further (2.85 [2.13–3.81]; P < 0.001). […]

CONCLUSIONS SDR alone, even without DKD, increases cardiovascular risk, particularly for PAD, independently of common cardiovascular risk factors in long-standing type 1 diabetes. More remains to be done to fully understand the link between SDR and CVD. This knowledge could help combat the enhanced cardiovascular risk beyond currently available regimens.”

“The 15-year cumulative incidence of any CVD in patients with and without SDR was 36.8% (95% CI 33.4–40.1) and 27.3% (23.3–31.0), respectively (P = 0.0004 for log-rank test) […] Patients without DKD and SDR at baseline had 4.0-fold (95% CI 3.3–4.7) increased risk of CVD compared with control subjects without diabetes up to 70 years of age […]. Intriguingly, after this age, the CVD incidence was similar to that in the matched control subjects (SIR 0.9 [95% CI 0.3–1.9]) in this subgroup of patients with diabetes. However, in patients without DKD but with SDR, the CVD risk was still increased after the patients had reached 70 years of age (SIR 3.4 [95% CI 1.8–6.2]) […]. Of note, in patients with both DKD and SDR, the CVD burden was high already at young ages.”

“This study highlights the role of SDR on a complete range of CVD outcomes in a large sample of patients with long-standing T1D and longitudinal follow-up. We showed that SDR alone, without concomitant DKD, increases the risk of macrovascular disease, independently of the traditional risk factors. The risk is further increased in case of accompanying DKD, especially if SDR is present together with DKD. Findings from this large and well-characterized cohort of patients have a direct impact on clinical practice, emphasizing the importance of regular screening for SDR in individuals with T1D and intensive multifactorial interventions for CVD prevention throughout their life span.

This study also confirms and complements previous data on the continuum of diabetic vascular disease, by which microvascular and macrovascular disease do not seem to be separate diseases, but rather interconnected (10,12,18). The link is most obvious for DKD, which clearly emerges as a major predictor of cardiovascular morbidity and mortality (2,24,25). The association of SDR with CVD is less clear. However, our recent cross-sectional study with the Joslin Medalist Study showed that the CVD risk was in fact increased in patients with SDR on top of DKD compared with DKD alone (19). In the present longitudinal study, we were able to extend those results also to show that SDR alone, without DKD and after the adjustment for other traditional risk factors, increases CVD risk substantially. SDR further increases CVD risk in case DKD is present as well. In addition, the role of SDR as an independent CVD risk predictor is also supported by our data using albuminuria as a marker of DKD. This is important because albuminuria is a known predictor of diabetic retinopathy progression (26) as well as a recognized biomarker for CVD.”

“A novel finding is that, independently of any signs of DKD, the risk of PAD is increased twofold in the presence of SDR. Although this association has recently been highlighted in individuals with type 2 diabetes (10,29), the data in T1D are scarce (16,30). Notably, the previous studies mostly lack adjustments for DKD, the major predictor of mortality in patients with shorter diabetes duration. Both complications, besides sharing some conventional cardiovascular risk factors, may be linked by additional pathological processes involving changes in the microvasculature in both the retina and the vasa vasorum of the conductance vessels (31). […] Patients with T1D duration of >30 years face a continuously increased CVD risk that is further increased by the occurrence of advanced PDR. Therefore, by examining the retina, additional insight into individual CVD risk is gained and can guide the clinician to a more tailored approach to CVD prevention. Moreover, our findings suggest that the link between SDR and CVD is at least partially independent of traditional risk factors, and the mechanism behind the phenomenon warrants further research, aiming to find new therapies to alleviate the CVD burden more efficiently.”

The model selection method employed in the paper is far from optimal [“Variables for the model were chosen based on significant univariable associations.” – This is not the way to do things!], but regardless these are interesting results.

iii. Fasting Glucose Variability in Young Adulthood and Cognitive Function in Middle Age: The Coronary Artery Risk Development in Young Adults (CARDIA) Study.

“Individuals with type 2 diabetes (T2D) have 50% greater risk for the development of neurocognitive dysfunction relative to those without T2D (13). The American Diabetes Association recommends screening for the early detection of cognitive impairment for adults ≥65 years of age with diabetes (4). Coupled with the increasing prevalence of prediabetes and diabetes, this calls for better understanding of the impact of diabetes on cerebral structure and function (5,6). Among older individuals with diabetes, higher intraindividual variability in glucose levels around the mean is associated with worse cognition and the development of Alzheimer disease (AD) (7,8). […] Our objectives were to characterize fasting glucose (FG) variability during young adulthood before the onset of diabetes and to assess whether such variability in FG is associated with cognitive function in middle adulthood. We hypothesized that a higher variability of FG during young adulthood would be associated with a lower level of cognitive function in midlife compared with lower FG variability.”

“We studied 3,307 CARDIA (Coronary Artery Risk Development in Young Adults) Study participants (age range 18–30 years and enrolled in 1985–1986) at baseline and calculated two measures of long-term glucose variability: the coefficient of variation about the mean FG (CV-FG) and the absolute difference between successive FG measurements (average real variability [ARV-FG]) before the onset of diabetes over 25 and 30 years of follow-up. Cognitive function was assessed at years 25 (2010–2011) and 30 (2015–2016) with the Digit Symbol Substitution Test (DSST), Rey-Auditory Verbal Learning Test (RAVLT), Stroop Test, Montreal Cognitive Assessment, and category and letter fluency tests. We estimated the association between glucose variability and cognitive function test score with adjustment for clinical and behavioral risk factors, mean FG level, change in FG level, and diabetes development, medication use, and duration.

RESULTS After multivariable adjustment, 1-SD increment of CV-FG was associated with worse cognitive scores at year 25: DSST, standardized regression coefficient −0.95 (95% CI −1.54, −0.36); RAVLT, −0.14 (95% CI −0.27, −0.02); and Stroop Test, 0.49 (95% CI 0.04, 0.94). […] We did not find evidence for effect modification by race or sex for any variability-cognitive function association”

CONCLUSIONS Higher intraindividual FG variability during young adulthood below the threshold of diabetes was associated with worse processing speed, memory, and language fluency in midlife independent of FG levels. […] In this cohort of black and white adults followed from young adulthood into middle age, we observed that greater intraindividual variability in FG below a diabetes threshold was associated with poorer cognitive function independent of behavioral and clinical risk factors. This association was observed above and beyond adjustment for concurrent glucose level; change in FG level during young adulthood; and diabetes status, duration, and medication use. Intraindividual glucose variability as determined by CV was more strongly associated with cognitive function than was absolute average glucose variability.”

iv. Maternal Antibiotic Use During Pregnancy and Type 1 Diabetes in Children — A National Prospective Cohort Study. It is important that papers like these get published and read, even if the results may not sound particularly exciting:

“Prenatal prescription of antibiotics is common but may perturb the composition of the intestinal microbiota in the offspring. In childhood the latter may alter the developing immune system to affect the pathogenesis of type 1 diabetes (1). Previous epidemiological studies reported conflicting results regarding the association between early exposure to antibiotics and childhood type 1 diabetes (2,3). Here we investigated the association in a Danish register setting.

The Danish National Birth Cohort (DNBC) provided data from 100,418 pregnant women recruited between 1996 and 2002 and their children born between 1997 and 2003 (n = 96,840). The women provided information on exposures during and after pregnancy. Antibiotic prescription during pregnancy was obtained from the Danish National Prescription Registry (anatomical therapeutic chemical code J01) [it is important to note that: “In Denmark, purchasing antibiotics requires a prescription, and all purchases are registered at the Danish National Prescription Registry”], and type 1 diabetes diagnoses (diagnostic codes DE10 and DE14) during childhood and adolescence were obtained from the Danish National Patient Register. The children were followed until 2014 (mean follow-up time 14.3 years [range 11.5–18.4 years, SD 1.4]).”

“A total of 336 children developed type 1 diabetes during follow-up. Neither overall exposure (hazard ratio [HR] 0.90; 95% CI 0.68–1.18), number of courses (HR 0.36–0.97[…]), nor trimester-specific exposure (HR 0.81–0.89 […]) of antibiotics in utero was associated with childhood diabetes. Moreover, exposure to specific types of antibiotics in utero did not change the risk of childhood type 1 diabetes […] This large prospective Danish cohort study demonstrated that maternal use of antibiotics during pregnancy was not associated with childhood type 1 diabetes. Thus, the results from this study do not support a revision of the clinical recommendations on treatment with antibiotics during pregnancy.”

v. Decreasing Cumulative Incidence of End-Stage Renal Disease in Young Patients With Type 1 Diabetes in Sweden: A 38-Year Prospective Nationwide Study.

“Diabetic nephropathy is a devastating complication to diabetes. It can lead to end-stage renal disease (ESRD), which demands renal replacement therapy (RRT) with dialysis or kidney transplantation. In addition, diabetic nephropathy is associated with increased risk of cardiovascular morbidity and mortality (1,2). As a nation, Sweden, next to Finland, has the highest incidence of type 1 diabetes in the world (3), and the incidence of childhood-onset diabetes is increasing globally (4,5). The incidence of ESRD caused by diabetic nephropathy in these Nordic countries is fairly low as shown in recent studies, 3–8% at maximum 30 years’ of diabetes duration (6,7). This is to be compared with studies from Denmark in the 1980s that showed a cumulative incidence of diabetic nephropathy of 41% at 40 years of diabetes duration. Older, hospital-based cohort studies found that the incidence of persistent proteinuria seemed to peak at 25 years of diabetes duration; after that, the incidence levels off (8,9). This implies the importance of genetic susceptibility as a risk factor for diabetic nephropathy, which has also been indicated in recent genome-wide scan studies (10,11). Still, modifiable factors such as metabolic control are clearly of major importance in the development of diabetic nephropathy (1215). Already in 1994, a decreasing incidence of diabetic nephropathy was seen in a hospital-based study in Sweden, and the authors concluded that this was mainly driven by better metabolic control (16). Young age at onset of diabetes has previously been found to protect, or postpone, the development of ESRD caused by diabetic nephropathy, while diabetes onset at older ages is associated with increased risk (7,9,17). In a previous study, we found that age at onset of diabetes affects men and women differently (7). Earlier studies have indicated a male predominance (8,18), while our previous study showed that the incidence of ESRD was similar in men and women with diabetes onset before 20 years of age, but with diabetes onset after 20 years of age, men had increased risk of developing ESRD compared with women. The current study analyzes the incidence of ESRD due to type 1 diabetes, and changes over time, in a large Swedish population-based cohort with a maximum follow-up of 38 years.”

“Earlier studies have shown that it takes ∼15 years to develop persistent proteinuria and another 10 to proceed to ESRD (9,25). In the current study population, no patients developed ESRD because of type 1 diabetes at a duration <14 years; thus only patients with diabetes duration of ≥14 years were included in the study. […] A total of 18,760 unique patients were included in the study: 10,560 (56%) men and 8,200 (44%) women. The mean age at the end of the study was somewhat lower for women, 38.9 years, compared with 40.2 years for men. Women tend to develop type 1 diabetes about a year earlier than men: mean age 15.0 years for women compared with 16.5 years for men. There was no difference regarding mean diabetes duration between men and women in the study (23.8 years for women and 23.7 years for men). A total of 317 patients had developed ESRD due to diabetes. The maximum diabetes duration was 38.1 years for patients in the SCDR and 32.6 years for the NDR and the DISS. The median time from onset of diabetes to ESRD was 22.9 years (minimum 14.1 and maximum 36.6). […] At follow-up, 77 patients with ESRD and 379 without ESRD had died […]. The risk of dying during the course of the study was almost 12 times higher among the ESRD patients (HR 11.9 [95% CI 9.3–15.2]) when adjusted for sex and age. Males had almost twice as high a risk of dying as female patients (HR 1.7 [95% CI 1.4–2.1]), adjusted for ESRD and age.”

“The overall incidence rate of ESRD during 445,483 person-years of follow-up was 0.71 per 1,000 person-years. […] The incidence rate increases with diabetes duration. For patients with diabetes onset at 0–9 and 10–19 years of age, there was an increase in incidence up to 36 years of duration; at longer durations, the number of cases is too small and results must be interpreted with caution. With diabetes onset at 20–34 years of age the incidence rate increases until 25 years of diabetes duration, and then a decrease can be observed […] In comparison of different time periods, the risk of developing ESRD was lower in patients with diabetes onset in 1991–2001 compared with onset in 1977–1984 (HR 3.5 [95% CI 2.3–5.3]) and 1985–1990 (HR 2.6 [95% CI 1.7–3.8]), adjusted for age at follow-up and sex. […] The lowest risk of developing ESRD was found in the group with onset of diabetes before the age of 10 years — both for males and females […]. With this group as reference, males diagnosed with diabetes at 10–19 or 20–34 years of age had increased risk of ESRD (HR 2.4 [95% CI 1.6–3.5] and HR 2.2 [95% CI 1.4–3.3]), respectively. For females, the risk of developing ESRD was also increased with diabetes onset at 10–19 years of age (HR 2.4 [95% CI 1.5–3.6]); however, when diabetes was diagnosed after the age of 20 years, the risk of developing ESRD was not increased compared with an early onset of diabetes (HR 1.4 [95% CI 0.8–3.4]).”

“By combining data from the SCDR, DISS, and NDR registers and identifying ESRD cases via the SRR, we have included close to all patients with type 1 diabetes in Sweden with diabetes duration >14 years who developed ESRD since 1991. The cumulative incidence of ESRD in this study is low: 5.6% (5.9% and 5.3% for males and females, respectively) at maximum 38 years of diabetes duration. For the first time, we could see a clear decrease in ESRD incidence in Sweden by calendar year of diabetes onset. The results are in line with a recent study from Norway that reported a modest incidence of 5.3% after 40 years of diabetes duration (27). In the current study, we found a decrease in the incidence rate after 25 years of diabetes duration in the group with diabetes onset at 20–34 years. With age at onset of diabetes 0–9 or 10–19 years, the ESRD incidence rate increases until 35 years of diabetes duration, but owing to the limited number of patients with longer duration we cannot determine whether the peak incidence has been reached or not. We can, however, conclude that the onset of ESRD has been postponed at least 10 years compared with that in older prospective cohort studies (8,9). […] In conclusion, this large population-based study shows a low incidence of ESRD in Swedish patients with onset of type 1 diabetes after 1977 and an encouraging decrease in risk of ESRD, which is probably an effect of improved diabetes care. We confirm that young age at onset of diabetes protects against, or prolongs, the time until development of severe complications.”

vi. Hypoglycemia and Incident Cognitive Dysfunction: A Post Hoc Analysis From the ORIGIN Trial. Another potentially important negative result, this one related to the link between hypoglycemia and cognitive impairment:

“Epidemiological studies have reported a relationship between severe hypoglycemia, cognitive dysfunction, and dementia in middle-aged and older people with type 2 diabetes. However, whether severe or nonsevere hypoglycemia precedes cognitive dysfunction is unclear. Thus, the aim of this study was to analyze the relationship between hypoglycemia and incident cognitive dysfunction in a group of carefully followed patients using prospectively collected data in the Outcome Reduction with Initial Glargine Intervention (ORIGIN) trial.”

“This prospective cohort analysis of data from a randomized controlled trial included individuals with dysglycemia who had additional cardiovascular risk factors and a Mini-Mental State Examination (MMSE) score ≥24 (N = 11,495). Severe and nonsevere hypoglycemic events were collected prospectively during a median follow-up time of 6.2 years. Incident cognitive dysfunction was defined as either reported dementia or an MMSE score of <24. The hazard of at least one episode of severe or nonsevere hypoglycemia for incident cognitive dysfunction (i.e., the dependent variable) from the time of randomization was estimated using a Cox proportional hazards model after adjusting for baseline cardiovascular disease, diabetes status, treatment allocation, and a propensity score for either form of hypoglycemia.

RESULTS This analysis did not demonstrate an association between severe hypoglycemia and incident cognitive impairment either before (hazard ratio [HR] 1.16; 95% CI 0.89, 1.52) or after (HR 1.00; 95% CI 0.76, 1.31) adjusting for the severe hypoglycemia propensities. Conversely, nonsevere hypoglycemia was inversely related to incident cognitive impairment both before (HR 0.59; 95% CI 0.52, 0.68) and after (HR 0.58; 95% CI 0.51, 0.67) adjustment.

CONCLUSIONS Hypoglycemia did not increase the risk of incident cognitive dysfunction in 11,495 middle-aged individuals with dysglycemia. […] These findings provide no support for the hypothesis that hypoglycemia causes long-term cognitive decline and are therefore reassuring for patients and their health care providers.”

vii. Effects of Severe Hypoglycemia on Cardiovascular Outcomes and Death in the Veterans Affairs Diabetes Trial.

“The VADT was a large randomized controlled trial aimed at determining the effects of intensive treatment of T2DM in U.S. veterans (9). In the current study, we examine predictors and consequences of severe hypoglycemia within the VADT and report several key findings. First, we identified risk factors for severe hypoglycemia that included intensive therapy, insulin use, proteinuria, and autonomic neuropathy. Consistent with prior reports in glucose-lowering studies, severe hypoglycemia occurred at a threefold significantly greater rate in those assigned to intensive glucose lowering. Second, severe hypoglycemia was associated with an increased risk of cardiovascular events, cardiovascular mortality, and all-cause mortality in both the standard and the intensive treatment groups. Of importance, however, severe hypoglycemia was associated with an even greater risk of all-cause mortality in the standard compared with the intensive treatment group. Third, the association between severe hypoglycemia and serious cardiovascular events was greater in individuals with an elevated risk for CVD at baseline.”

“Mean participant characteristics were as follows: age, 60.4 years; duration of diabetes, 11.5 years; BMI, 31.3 kg/m2; and HbA1c, 9.4%. Seventy-two percent had hypertension, 40% had a previous cardiovascular event, 62% had a microvascular complication, and 52% had baseline insulin use. The standard and intensive treatment groups included 899 and 892 participants, respectively. […] During the study, the standard treatment group averaged 3.7 severe hypoglycemic events per 100 patient-years versus 10.3 events per 100 patient-years in the intensive treatment group (P < 0.001). Overall, the combined rate of severe hypoglycemia during follow-up in the VADT from both study arms was 7.0 per 100 patient-years. […] Severe hypoglycemia within the prior 3 months was associated with an increased risk for composite cardiovascular outcome (HR 1.9 [95% CI 1.1, 3.5]; P = 0.03), cardiovascular mortality (3.7 [1.3, 10.4]; P = 0.01), and all-cause mortality (2.4 [1.1, 5.1]; P = 0.02) […]. More distant hypoglycemia (4–6 months prior) had no independently associated increased risk with adverse events or death. The association of severe hypoglycemia with cardiovascular events or cardiovascular mortality were not significantly different between the intensive and standard treatment groups […]. In contrast, the association of severe hypoglycemia with all-cause mortality was significantly greater in the standard versus the intensive treatment group (6.7 [2.7, 16.6] vs. 0.92 [0.2, 3.8], respectively; P = 0.019 for interaction). Because of the relative paucity of repeated severe hypoglycemic events in either study group, there was insufficient power to determine whether more than one episode of severe hypoglycemia increased the risk of subsequent outcomes.”

“Although recent severe hypoglycemia increased the risk of major cardiovascular events for those with a 10-year cardiovascular risk score of 35% (HR 2.88 [95% CI 1.57, 5.29]; absolute risk increase per 10 episodes = 0.252; number needed to harm = 4), hypoglycemia was not significantly associated with increased major cardiovascular events for those with a risk score of ≤7.5%. The absolute associated risk of major adverse cardiovascular events, cardiovascular mortality, and all-cause mortality increased with higher CVD risk for all three outcomes […]. We were not able to identify, however, any group of patients in either treatment arm in which severe hypoglycemia did not increase the risk of CVD events and mortality at least to some degree.”

“Although the explanation for the relatively greater risk of serious adverse events after severe hypoglycemia in the standard treatment group is unknown, we agree with previous reports that milder episodes of hypoglycemia, which are more frequent in the intensive treatment group, may quantitatively blunt the release of neuroendocrine and autonomic nervous system responses and their resultant metabolic and cardiovascular responses to hypoglycemia, thereby lessening the impact of subsequent severe hypoglycemic episodes (18,19). Episodes of prior hypoglycemia have rapid and significant effects on reducing (i.e., blunting) subsequent counterregulatory responses to a falling plasma glucose level (20,21). Thus, if one of the homeostatic counterregulatory responses (e.g., epinephrine) also can initiate unwanted intravascular atherothrombotic consequences, it may follow that severe hypoglycemia in a more intensively treated and metabolically well-controlled individual would provoke a reduced counterregulatory response. Although hypoglycemia frequency may be increased in these individuals, this may also lower unwanted and deleterious effects on the vasculature from counterregulatory responses. On the other hand, an isolated severe hypoglycemic event in a less well-controlled individual could provoke a relatively greater counterregulatory response with a proportionally attendant elevated risk for adverse vascular effects (22). In support of this, we previously reported in a subset of VADT participants that despite more frequent serious hypoglycemia in the intensive therapy group, progression of coronary artery calcium scores after severe hypoglycemia only occurred in the standard treatment group (23).”

“In the current study, we demonstrate that the association of severe hypoglycemia with subsequent serious adverse cardiovascular events and death occurred within the preceding 3 months but not beyond. The temporal relationship and proximity of severe hypoglycemia to a subsequent serious cardiovascular event and/or death has been investigated in a number of recent clinical trials in T2DM (25,13,14). All these trials consistently reported an association between severe hypoglycemic and subsequent serious adverse events. However, the proximity of severe hypoglycemic events to subsequent adverse events and death varies. In ADVANCE, a severe hypoglycemic episode increased the risk of major cardiovascular events for both the next 3 months and the following 6 months. In A Trial Comparing Cardiovascular Safety of Insulin Degludec Versus Insulin Glargine in Subjects With Type 2 Diabetes at High Risk of Cardiovascular Events (DEVOTE) and the Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results (LEADER) trial, there was an increased risk of either serious cardiovascular events or all-cause mortality starting 15 days and extending (albeit with decreasing risk) up to 1 year after severe hypoglycemia (13,14).”

June 15, 2019 Posted by | Cardiology, Diabetes, Epidemiology, Genetics, Nephrology, Neurology, Ophthalmology, Studies | Leave a comment

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

Oncology (II)

Here’s my first post in this series. Below some more quotes and links related to the book’s coverage.

Types of Pain
1. Nociceptive pain
a. Somatic pain: Cutaneous or musculoskeletal tissue (ie, bone, soft tissue metastases). Usually well-localized, increased w/use/movement.
b. Visceral pain: Compression, obstruction, infiltration, ischemia, stretching, or inflammation of solid & hollow viscera. Diffuse, nonfocal.
2. Neuropathic pain: Direct injury/dysfunction of peripheral or CNS tissues. Typically burning, radiating, may increase at rest or w/nerve stretching.
Pain emergencies: Pain crisis, spinal cord compression, fracture, bowel obstruction, severe mucositis, acute severe side effects of opioids (addiction crisis, delirium, respiratory depression), severe pain in imminently dying pt [patient, US]
Pain mgmt at the end of life is a moral obligation to alleviate pain & unnecessary suffering & is not euthanasia. (Vacco vs. Quill, U.S. Supreme Court, 1997)”

Nausea and Vomiting
Chemotherapy-induced N/V — 3 distinct types: Acute, delayed, & anticipatory. Acute begins 1–2 h after chemotherapy & peaks at 4–6 h, delayed begins at 24 h & peaks at 48–72 h, anticipatory is conditioned response to nausea a/w previous cycles of chemotherapy”

Constipation […] affects 50% of pts w/advanced CA; majority of pts being treated w/opioid analgesics, other contributors: malignant obstruction, ↓ PO/fluid intake, inactivity, anticholinergics, electrolyte derangement”

Fatigue
Prevalence/screening — occurs in up to 75% of all solid tumor pts & up to 99% of CA pts receiving multimodality Rx. Providers should screen for fatigue at initial visit, at dx [diagnosis, US] of advanced dz [disease] & w/each chemo visit; should assess for depression & insomnia w/new dx of fatigue (JCO 2008;23:3886) […] Several common 2° causes to eval & target include anemia (most common), thyroid or adrenal insufficiency, hypogonadism”

Delirium
*Definition — disturbances in level of consciousness, attention, cognition, and/or perception developing abruptly w/fluctuations over course of d *Clinical subtypes — hyperactive, hypoactive, & mixed […] *Maximize nonpharm intervention prior to pharmacology […] *Use of antipsychotics should be geared toward short-term use for acute sx [symptoms, US] *Benzodiazepines should only be initiated for delirium as an adjunct to antipsychotics in setting of agitation despite adequate antipsychotic dosing (JCO 2011;30:1206)”

Cancer Survivorship
Overview *W/improvement in dx & tx of CA, there are millions of CA survivors, & this number is increasing
*Pts experience the normal issues of aging, w/c are compounded by the long-term effects of CA & CA tx
*CA survivors are at ↑ risk of developing morbidity & illnesses at younger age than general population due to their CA tx […] ~312,570 male & ~396,080 female CA survivors <40 y of age (Cancer Treatment and Survivorship Facts and Figures 2016–2017, ACS) *Fertility is an important issue for survivors & there is considerable concern about the possibility of impairment (Human Reproduction Update 2009;15:587)”

“Pts undergoing cancer tx are at ↑ risk for infxn [infection, US] due to disease itself or its therapies. […] *Epidemiology: 10–50% of pts w/ solid tumors & >80% of pts with hematologic tumors *Source of infxn evident in only 20–30% of febrile episodes *If identified, common sites of infxn include blood, lungs, skin, & GI tract *Regardless of microbiologic diagnosis, Rx should be started within 2 h of fever onset which improves outcomes […] [Infections in the transplant host is the] Primary cause of death in 8% of auto-HCT & up to 20% of allo-HCT recipients” [here’s a relevant link, US].

Localized prostate cancer
*Epidemiology Incidence: ~180000, most common non-skin CA (2016: U.S. est.) (CA Cancer J Clin 2016:66:7) *Annual Mortality: ~26000, 2nd highest cause of cancer death in men (2016: U.S. est) […] Mortality benefit from screening asx [asymptomatic, US] men has not been definitively established, & individualized discussion of potential benefits & harms should occur before PSA testing is offered. […] Gleason grade reflects growth/differentiation pattern & ranges from 1–5, from most to least differentiated. […] Historical (pre-PSA) 15-y prostate CA mortality risk for conservatively managed (no surgery or RT) localized Gleason 6: 18–30%, Gleason 7: 42–70%, Gleason 8–10: 60–87% (JAMA 1998:280:975)”

Bladder cancer […] Most common malignancy of the urinary system, ~77000 Pts will be diagnosed in the US in 2016, ~16000 will die of their dz. […] Presenting sx: Painless hematuria (typically intermittent & gross), irritative voiding sx (frequency, urgency, dysuria), flank or suprapubic pain (symptomatic of locally advanced dz), constitutional sx (fatigue, wt loss, failure to thrive) usually symptomatic of met [metastatic, US] dz

Links:

WHO analgesia ladder. (But see also this – US).
Renal cell carcinoma (“~63000 new cases & ~1400 deaths in the USA in 2016 […] Median age dx 64, more prevalent in men”)
Germ cell tumour (“~8720 new cases of testicular CA in the US in 2016 […] GCT is the most common CA in men ages of 15 to 35 y/o”)
Non-small-cell lung carcinoma (“225K annual cases w/ 160K US deaths, #1 cause of cancer mortality; 70% stage III/IV *Cigarette smoking: 85% of all cases, ↑ w/ intensity & duration of smoking”)
Small-cell lung cancer. (“SCLC accounts for 13–15% of all lung CAs, seen almost exclusively in smokers, majority w/ extensive stage dz at dx (60–70%). Lambert–Eaton myasthenic syndrome (“Affects 3% of SCLC pts”).
Thymoma. Myasthenia gravis. Morvan’s syndrome. Masaoka-Koga Staging system.
Pleural mesothelioma (“Rare; ≅3000 new cases dx annually in US. Commonly develops in the 5th to 7th decade […] About 80% are a/w asbestos exposure. […] Develops decades after asbestos exposure, averaging 30–40 years […] Median survival: 10 mo. […] Screening has not been shown to ↓ mortality even in subjects w/ asbestos exposure”)
Hepatocellular Carcinoma (HCC). (“*6th most common CA worldwide (626,000/y) & 2nd leading cause of worldwide CA mortality (598,000/y) *>80% cases of HCC occur in sub-Saharan Africa, eastern & southeastern Asia, & parts of Oceania including Papua New Guinea *9th leading cause of CA mortality in US […] Viral hepatitis: HBV & HCV are the leading RFs for HCC & accounts for 75% cases worldwide […] While HCV is now the leading cause of HCC in the US, NASH is expected to become a risk factor of increasing importance in the next decade”). Milan criteria.
CholangiocarcinomaKlatskin tumor. Gallbladder cancer. Courvoisier’s sign.
Pancreatic cancer (Incidence: estimated ~53,070 new cases/y & ~42,780 D/y in US (NCI SEER); 4th most common cause of CA death in US men & women; estimated to be 2nd leading cause of CA-related mortality by 2020″). Trousseau sign of malignancy. Whipple procedure.

October 21, 2018 Posted by | Books, Cancer/oncology, Gastroenterology, Medicine, Nephrology, Neurology, Psychiatry | Leave a comment

Circadian Rhythms (II)

Below I have added some more observations from the book, as well as some links of interest.

“Most circadian clocks make use of a sun-based mechanism as the primary synchronizing (entraining) signal to lock the internal day to the astronomical day. For the better part of four billion years, dawn and dusk has been the main zeitgeber that allows entrainment. Circadian clocks are not exactly 24 hours. So to prevent daily patterns of activity and rest from drifting (freerunning) over time, light acts rather like the winder on a mechanical watch. If the clock is a few minutes fast or slow, turning the winder sets the clock back to the correct time. Although light is the critical zeitgeber for much behaviour, and provides the overarching time signal for the circadian system of most organisms, it is important to stress that many, if not all cells within an organism possess the capacity to generate a circadian rhythm, and that these independent oscillators are regulated by a variety of different signals which, in turn, drive countless outputs […]. Colin Pittendrigh was one of the first to study entrainment, and what he found in Drosophila has been shown to be true across all organisms, including us. For example, if you keep Drosophila, or a mouse or bird, in constant darkness it will freerun. If you then expose the animal to a short pulse of light at different times the shifting (phase shifting) effects on the freerunning rhythm vary. Light pulses given when the clock ‘thinks’ it is daytime (subjective day) will have little effect on the clock. However, light falling during the first half of the subjective night causes the animal to delay the start of its activity the following day, while light exposure during the second half of the subjective night advances activity onset. Pittendrigh called this the ‘phase response curve’ […] Remarkably, the PRC of all organisms looks very similar, with light exposure around dusk and during the first half of the night causing a delay in activity the next day, while light during the second half of the night and around dawn generates an advance. The precise shape of the PRC varies between species. Some have large delays and small advances (typical of nocturnal species) while others have small delays and big advances (typical of diurnal species). Light at dawn and dusk pushes and pulls the freerunning rhythm towards an exactly 24-hour cycle. […] Light can act directly to modify behaviour. In nocturnal rodents such as mice, light encourages these animals to seek shelter, reduce activity, and even sleep, while in diurnal species light promotes alertness and vigilance. So circadian patterns of activity are not only entrained by dawn and dusk but also driven directly by light itself. This direct effect of light on activity has been called ‘masking’, and combines with the predictive action of the circadian system to restrict activity to that period of the light/dark cycle to which the organism has evolved and is optimally adapted.”

“[B]irds, reptiles, amphibians, and fish (but not mammals) have ‘extra-ocular’ photoreceptors located within the pineal complex, hypothalamus, and other areas of the brain, and like the invertebrates, eye loss in many cases has little impact upon the ability of these animals to entrain. […] Mammals are strikingly different from all other vertebrates as they possess photoreceptor cells only within their eyes. Eye loss in all groups of mammals […] abolishes the capacity of these animals to entrain their circadian rhytms to the light/dark cycle. But astonishingly, the visual cells of the retina – the rods and cones – are not required for the detection of the dawn/dusk signal. There exists a third class of photoreceptors within the eye […] Studies in the late 1990s by Russell Foster and his colleagues showed that mice lacking all their rod and cone photoreceptors could still regulate their circadian rhythms to light perfectly normally. But when their eyes were covered the ability to entrain was lost […] work on the rodless/coneless mouse, along with [other] studies […], clearly demonstrated that the mammalian retina contains a small population of photosensitive retinal ganglion cells or pRGCs, which comprise approximately 1-2 per cent of all retinal ganglion cells […] Ophthalmologists now appreciate that eye loss deprives us of both vision and a proper sense of time. Furthermore, genetic diseases that result in the loss of the rods and cones and cause visual blindness, often spare the pRGCs. Under these circumstances, individuals who have their eyes but are visually blind, yet possess functional pRGCs, need to be advised to seek out sufficient light to entrain their circadian system. The realization that the eye provides us with both our sense of space and our sense of time has redefined the diagnosis, treatment, and appreciation of human blindness.”

“But where is ‘the’ circadian clock of mammals? […] [Robert] Moore and [Irving] Zucker’s work pinpointed the SCN as the likely neural locus of the light-entrainable circadian pacemaker in mammals […] and a decade later this was confirmed by definitive experiments from Michael Menaker’s laboratory undertaken at the University of Virginia. […] These experiments established the SCN as the ‘master circadian pacemaker’ of mammals. […] There are around 20,000 or so neurons in the mouse SCN, but they are not identical. Some receive light information from the pRGCs and pass this information on to other SCN neurons, while others project to the thalamus and other regions of the brain, and collectively these neurons secrete more than one hundred different neurotransmitters, neuropeptides, cytokines, and growth factors. The SCN itself is composed of several regions or clusters of neurons, which have different jobs. Furthermore, there is considerable variability in the oscillations of the individual cells, ranging from 21.25 to 26.25 hours. Although the individual cells in the SCN have their own clockwork mechanisms with varying periods, the cell autonomous oscillations in neural activity are synchronized at the system level within the SCN, providing a coherent near 24-hour signal to the rest of the mammal. […] SCN neurons exhibit a circadian rhythm of spontaneous action potentials (SAPs), with higher frequency during the daytime than the night which in turn drives many rhythmic changes by alternating stimulatory and inhibitory inputs to the appropriate target neurons in the brain and neuroendocrine systems. […] The SCN projects directly to thirty-five brain regions, mostly located in the hypothalamus, and particularly those regions of the hypothalamus that regulate hormone release. Indeed, many pituitary hormones, such as cortisol, are under tight circadian control. Furthermore, the SCN regulates the activity of the autonomous nervous system, which in turn places multiple aspects of physiology, including the sensitivity of target tissues to hormonal signals, under circadian control. In addition to these direct neuronal connections, the SCN communicates to the rest of the body using diffusible chemical signals.”

“The SCN is the master clock in mammals but it is not the only clock. There are liver clocks, muscle clocks, pancreas clocks, adipose tissue clocks, and clocks of some sort in every organ and tissue examined to date. While lesioning of the SCN disrupts global behavioural rhythms such as locomotor activity, the disruption of clock function within just the liver or lung leads to circadian disorder that is confined to the target organ. In tissue culture, liver, heart, lung, skeletal muscle, and other organ tissues such as mammary glands express circadian rhythms, but these rhythms dampen and disappear after only a few cycles. This occurs because some individual clock cells lose rhythmicity, but more commonly because the individual cellular clocks become uncoupled from each other. The cells continue to tick, but all at different phases so that an overall 24-hour rhythm within the tissue or organ is lost. The discovery that virtually all cells of the body have clocks was one of the big surprises in circadian rhythms research. […] the SCN, entrained by pRGCs, acts as a pacemaker to coordinate, but not drive, the circadian activity of billions of individual peripheral circadian oscillators throughout the tissues and organs of the body. The signalling pathways used by the SCN to phase-entrain peripheral clocks are still uncertain, but we know that the SCN does not send out trillions of separate signals around the body that target specific cellular clocks. Rather there seems to be a limited number of neuronal and humoral signals which entrain peripheral clocks that in turn time their local physiology and gene expression.”

“As in Drosophilia […], the mouse clockwork also comprises three transcriptional-translational feedback loops with multiple interacting components. […] [T]he generation of a robust circadian rhythm that can be entrained by the environment is achieved via multiple elements, including the rate of transcription, translation, protein complex assembly, phosphorylation, other post-translation modification events, movement into the nucleus, transcriptional inhibition, and protein degradation. […] [A] complex arrangement is needed because from the moment a gene is switched on, transcription and translation usually takes two hours at most. As a result, substantial delays must be imposed at different stages to produce a near 24-hour oscillation. […] Although the molecular players may differ from Drosophilia and mice, and indeed even between different insects, the underlying principles apply across the spectrum of animal life. […] In fungi, plants, and cyanobacteria the clock genes are all different from each other and different again from the animal clock genes, suggesting that clocks evolved independently in the great evolutionary lineages of life on earth. Despite these differences, all these clocks are based upon a fundamental TTFL.”

“Circadian entrainment is surprisingly slow, taking several days to adjust to an advanced or delayed light/dark cycle. In most mammals, including jet-lagged humans, behavioural shifts are limited to approximately one hour (one time zone) per day. […] Changed levels of PER1 and PER2 act to shift the molecular clockwork, advancing the clock at dawn and delaying the clock at dusk. However, per mRNA and PER protein levels fall rapidly even if the animal remains exposed to light. As a result, the effects of light on the molecular clock are limited and entrainment is a gradual process requiring repeated shifting stimuli over multiple days. This phenomenon explains why we get jet lag: the clock cannot move immediately to a new dawn/dusk cycle because there is a ‘brake’ on the effects of light on the clock. […] The mechanism that provides this molecular brake is the production of SLK1 protein. […] Experiments on mice in which SLK1 has been suppressed show very rapid entrainment to simulated jet-lag.”

“We spend approximately 36 per cent of our entire lives asleep, and while asleep we do not eat, drink, or knowingly pass on our genes. This suggests that this aspect of our 24-hour behaviour provides us with something of huge value. If we are deprived of sleep, the sleep drive becomes so powerful that it can only be satisfied by sleep. […] Almost all life shows a 24-hour pattern of activity and rest, as we live on a planet that revolves once every 24 hours causing profound changes in light, temperature, and food availability. […] Life seems to have made an evolutionary ‘decision’ to be active at a specific part of the day/night cycle, and a species specialized to be active during the day will be far less effective at night. Conversely, nocturnal animals that are beautifully adapted to move around and hunt under dim or no light fail miserably during the day. […] no species can operate with the same effectiveness across the 24-hour light/dark environment. Species are adapted to a particular temporal niche just as they are to a physical niche. Activity at the wrong time often means death. […] Sleep may be the suspension of most physical activity, but a huge amount of essential physiology occurs during this time. Many diverse processes associated with the restoration and rebuilding of metabolic pathways are known to be up-regulated during sleep […] During sleep the body performs a broad range of essential ‘housekeeping’ functions without which performance and health during the active phase deteriorates rapidly. But these housekeeping functions would not be why sleep evolved in the first place. […] Evolution has allocated these key activities to the most appropriate time of day. […] In short, sleep has probably evolved as a species-specific response to a 24-hour world in which light, temperature, and food availability change dramatically. Sleep is a period of physical inactivity when individuals avoid movement within an environment to which they are poorly adapted, while using this time to undertake essential housekeeping functions demanded by their biology.”

“Sleep propensity in humans is closely correlated with the melatonin profile but this may be correlation and not causation. Indeed, individuals who do not produce melatonin (e.g. tetraplegic individuals, people on beta-blockers, or pinealectomized patients) still exhibit circadian sleep/wake rhythms with only very minor detectable changes. Another correlation between melatonin and sleep relates to levels of alertness. When melatonin is suppressed by light at night alertness levels increase, suggesting that melatonin and sleep propensity are directly connected. However, increases in alertness occur before a significant drop in blood melatonin. Furthermore, increased light during the day will also improve alertness when melatonin levels are already low. These findings suggest that melatonin is not a direct mediator of alertness and hence sleepiness. Taking synthetic melatonin or synthetic analogues of melatonin produces a mild sleepiness in about 70 per cent of people, especially when no natural melatonin is being released. The mechanism whereby melatonin produces mild sedation remains unclear.”

Links:

Teleost multiple tissue (tmt) opsin.
Melanopsin.
Suprachiasmatic nucleus.
Neuromedin S.
Food-entrainable circadian oscillators in the brain.
John Harrison. Seymour Benzer. Ronald Konopka. Jeffrey C. Hall. Michael Rosbash. Michael W. Young.
Circadian Oscillators: Around the Transcription-Translation Feedback Loop and on to Output.
Period (gene). Timeless (gene). CLOCK. Cycle (gene). Doubletime (gene). Cryptochrome. Vrille Gene.
Basic helix-loop-helix.
The clockwork orange Drosophila protein functions as both an activator and a repressor of clock gene expression.
RAR-related orphan receptor. RAR-related orphan receptor alpha.
BHLHE41.
The two-process model of sleep regulation: a reappraisal.

September 30, 2018 Posted by | Books, Genetics, Medicine, Molecular biology, Neurology, Ophthalmology | Leave a comment

100 Cases in Orthopaedics and Rheumatology (II)

Below I have added some links related to the last half of the book’s coverage, as well as some more observations from the book.

Scaphoid fracture. Watson’s test. Dorsal intercalated segment instability. (“Non-union is not uncommon as a complication after scaphoid fractures because the blood supply to this bone is poor. Smokers have a higher incidence of non-union. Occasionally, the blood supply is poor enough to lead to avascular necrosis. If non-union is not detected, subsequent arthritis in the wrist can develop.”)
Septic arthritis. (“Septic arthritis is an orthopaedic emergency. […] People with septic arthritis are typically unwell with fevers and malaise and the joint pain is severe. […] Any acutely hot or painful joint is septic arthritis until proven otherwise.”)
Rheumatoid arthritis. (“[RA is] the most common of the inflammatory arthropathies. […] early-morning stiffness and pain, combined with soft-tissue rather than bony swelling, are classic patterns for inflammatory disease. Although […] RA affects principally the small joints of the hands (and feet), it may progress to involve any synovial joint and may be complicated by extra-articular features […] family history [of the disease] is not unusual due to the presence of susceptibility genes such as HLA-DR. […] Not all patients with RA have rheumatoid factor (RF), and not all patients with RF have RA; ACPA has greater specificity for RA than rheumatoid factor. […] Medical therapy focuses on disease-modifying anti-rheumatic drugs (DMARDs) such as methotrexate, sulphasalazine, leflunomide and hydroxychloroquine which may be used individually or in combination. […] Disease activity in RA is measured by the disease activity score (DAS), which is a composite score of the clinical evidence of synovitis, the current inflammatory response and the patient’s own assessment of their health. […] Patients who have high disease activity as determined by the DAS and have either failed or failed to tolerate standard disease modifying therapy qualify for biologic therapy – monoclonal antibodies that are directed against key components of the inflammatory response. […] TNF-α blockade is highly effective in up to 70 per cent of patients, reducing both inflammation and the progressive structural damage associated with severe active disease.”)
Ankylosing spondylitis. Ankylosis. Schober’s index. Costochondritis.
Mononeuritis multiplex. (“Mononeuritis multiplex arises due to interruption of the vasa nervorum, the blood supply to peripheral nerves […] Mononeuritis multiplex is commonly caused by diabetes or vasculitis. […] Vasculitis – inflammation of blood vessels and subsequent obstruction to blood flow – can be primary (idiopathic) or secondary, in which case it is associated with an underlying condition such as rheumatoid arthritis. The vasculitides are classified according to the size of the vessel involved. […] Management of mononeuritis multiplex is based on potent immunosuppression […] and the treatment of underlying infections such as hepatitis.”)
Multiple myeloma. Bence-Jones protein. (“The combination of bone pain and elevated ESR and calcium is suggestive of multiple myeloma.”)
Osteoporosis. DEXA scan. T-score. (“Postmenopausal bone loss is the most common cause of osteoporosis, but secondary osteoporosis may occur in the context of a number of medical conditions […] Steroid-induced osteoporosis is a significant problem in medical practice. […] All patients receiving corticosteroids should have bone protection […] Pharmacological treatment in the form of calcium supplementation and biphosphonates to reduce osteoclast activity is effective but compliance is typically poor.”)
Osteomalacia. Rickets. Craniotabes.
Paget’s disease (see also this post). (“In practical terms, the main indication to treat Paget’s disease is pain […] although bone deformity or compression syndromes (or risk thereof) would also prompt therapy. The treatment of choice is a biphosphonate to diminish osteoclast activity”).
Stress fracture. Female athlete triad. (“Stress fractures are overuse injuries and occur when periosteal resorption exceeds bone formation. They are commonly seen in two main patient groups: soldiers may suffer so-called march fractures in the metatarsals, while athletes may develop them in different sites according to their sporting activity. Although the knee is a common site in runners due to excess mechanical loading, stress fractures may also result in non-weight-bearing sites due to repetitive and excessive traction […]. The classic symptom […] is of pain that occurs throughout running and crucially persists with rest; this is in contrast to shin splints, a traction injury to the tibial periosteum in which the pain diminishes somewhat with continued activity […] The crucial feature of rehabilitation is a graded return to sport to prevent progression or recurrence.”)
Psoriatic arthritis. (“Arthropathy and rash is a common combination in rheumatology […] Psoriatic arthritis is a common inflammatory arthropathy that affects up to 15 per cent of those with psoriasis. […] Nail disease is very helpful in differentiating psoriatic arthritis from other forms of inflammatory arthropathy.”)
Ehlers–Danlos syndromes. Marfan syndrome. Beighton (hypermobility) score.
Carpal tunnel syndrome. (“Carpal tunnel syndrome is the most common entrapment neuropathy […] The classic symptoms are of tingling in the sensory distribution of the median nerve (i.e. the lateral three and a half digits); loss of thumb abduction is a late feature. Symptoms are often worse at night (when the hand might be quite painful) and in certain postures […] The majority of cases are idiopathic, but pregnancy and rheumatoid arthritis are very common precipitating causes […] The majority of patients will respond well to conservative management […] If these measures fail, corticosteroid injection into the carpal tunnel can be very effective in up to 80 per cent of patients. Surgical decompression should be reserved for those with persistent disabling symptoms or motor loss.”)
Mixed connective tissue disease.
Crystal arthropathy. Tophus. Uric acid nephropathyChondrocalcinosis. (“In any patient presenting with an acutely painful and swollen joint, the most important diagnoses to consider are septic arthritis and crystal arthropathy. Crystal arthropathy such as gout is more common than septic arthritis […] Gout may be precipitated by diuretics, renal impairment and aspirin use”).
Familial Mediterranean fever. Amyloidosis.
Systemic lupus erythematosus (see also this). Jaccoud arthropathy. Lupus nephritis. (“Renal disease is the most feared complication of SLE.”)
Scleroderma. Raynaud’s phenomenon. (“Scleroderma is an uncommon disorder characterized by thickening of the skin and, to a greater or lesser degree, fibrosis of internal organs.”)
Henoch-Schönlein purpura. Cryoglobulinemia. (“Purpura are the result of a spontaneous extravasation of blood from the capillaries into the skin. If small they are known as petechiae, when they are large they are termed ecchymoses. There is an extensive differential diagnosis for purpura […] The combination of palpable purpura (distributed particularly over the buttocks and extensor surfaces of legs), abdominal pain, arthritis and renal disease is a classic presentation of Henoch–Schönlein purpura (HSP). HSP is a distinct and frequently self-limiting small-vessel vasculitis that can affect any age; but the majority of cases present in children aged 2–10 years, in whom the prognosis is more benign than the adult form, often remitting entirely within 3–4 months. The abdominal pain may mimic a surgical abdomen and can presage intussusception, haemorrhage or perforation. The arthritis, in contrast, is relatively mild and tends to affect the knees and ankles.”)
Rheumatic fever.
Erythema nodosum. (“Mild idiopathic erythema nodosum […] needs no specific treatment”).
Rheumatoid lung disease. Bronchiolitis obliterans. Methotrexate-induced pneumonitis. Hamman–Rich syndrome.
Antiphospholipid syndrome. Sapporo criteria. (“Antiphospholipid syndrome is a hypercoagulable state characterized by recurrent arteriovenous thrombosis and/or pregnancy morbidity in the presence of either a lupus anticoagulant or anticardiolipin antibody (both phospholipid-related proteins). […] The most common arteriovenous thrombotic events in antiphospholipid syndrome are deep venous thrombosis and pulmonary embolus […], but any part of the circulation may be involved, with arterial events such as myocardial infarction and stroke carrying a high mortality rate. Poor placental circulation is thought to be responsible for the high pregnancy morbidity, with recurrent first- and second-trimester loss and a higher rate of pre-eclampsia being typical clinical features.”)
Still’s disease. (“Consider inflammatory disease in cases of pyrexia of unknown origin.”)
Polymyalgia rheumatica. Giant cell arteritis. (“[P]olymyalgia rheumatica (PMR) [is] a systemic inflammatory syndrome affecting the elderly that is characterized by bilateral pain and stiffness in the shoulders and hip girdles. The stiffness can be profound and limits mobility although true muscle weakness is not a feature. […] The affected areas are diffusely tender, with movements limited by pain. […] care must be taken not to attribute joint inflammation to PMR until other diagnoses have been excluded; for example, a significant minority of RA patients may present with a polymyalgic onset. […] The treatment for PMR is low-dose corticosteroids. […] Many physicians would consider a dramatic response to low-dose prednisolone as almost diagnostic for PMR, so if a patients symptoms do not improve rapidly it is wise to re-evaluate the original diagnosis.”)
Relapsing polychondritis. (“Relapsing polychondritis is characterized histologically by inflammatory infiltration and later fibrosis of cartilage. Any cartilage, in any location, is at risk. […] Treatment of relapsing polychondritis is with corticosteroids […] Surgical reconstruction of collapsed structures is not an option as the deformity tends to continue postoperatively.”)
Dermatomyositis. Gottron’s Papules.
Enteropathic arthritis. (“A seronegative arthritis may develop in up to 15 per cent of patients with any form of inflammatory bowel disease, including ulcerative colitis (UC), Crohn’s disease or microscopic and collagenous colitis. The most common clinical presentations are a peripheral arthritis […] and spondyloarthritis.”)
Reflex sympathetic dystrophy.
Whipple’s disease. (“Although rare, consider Whipple’s disease in any patient presenting with malabsorption, weight loss and arthritis.”)
Wegener’s granulomatosis. (“Small-vessel vasculitis may cause a pulmonary-renal syndrome. […] The classic triad of Weneger’s granulomatosis is the presence of upper and lower respiratory tract disease and renal impairment.”)
Reactive arthritis. Reiter’s syndrome. (“Consider reactive arthritis in any patient presenting with a monoarthropathy. […] Reactive arthritis is generally benign, with up to 80 per cent making a full recovery.”)
Sarcoidosis. Löfgren syndrome.
Polyarteritis nodosa. (“Consider mesenteric ischaemia in any patient presenting with a systemic illness and postprandial abdominal pain.”)
Sjögren syndrome. Schirmer’s test.
Behçet syndrome.
Lyme disease. Erythema chronicum migrans. (“The combination of rash leading to arthralgia and cranial neuropathy is a classic presentation of Lyme disease.”)
Takayasu arteritis. (“Takayasu’s arteritis is an occlusive vasculitis leading to stenoses of the aorta and its principal branches. The symptoms and signs of the disease depend on the distribution of the affected vessel but upper limbs are generally affected more commonly than the iliac tributaries. […] the disease is a chronic relapsing and remitting condition […] The mainstay of treatment is high-dose corticosteroids plus a steroid-sparing agent such as methotrexate. […] Cyclophosphamide is reserved for those patients who do not achieve remission with standard therapy. Surgical intervention such as bypass or angioplasty may improve ischaemic symptoms once the inflammation is under control.”)
Lymphoma.
Haemarthrosis. (“Consider synovial tumours in a patient with unexplained haemarthrosis.”)
Juvenile idiopathic arthritis.
Drug-induced lupus erythematosus. (“Drug-induced lupus (DIL) generates a different spectrum of clinical manifestations from idiopathic disease. DIL is less severe than idiopathic SLE, and nephritis or central nervous system involvement is very rare. […] The most common drugs responsible for a lupus-like syndrome are procainamide, hydralazine, quinidine, isoniazid, methyldopa, chlorpromazine and minocycline. […] Treatment involves stopping the offending medication and the symptoms will gradually resolve.”)
Churg–Strauss syndrome.

July 8, 2018 Posted by | Books, Cancer/oncology, Cardiology, Gastroenterology, Immunology, Medicine, Nephrology, Neurology, Ophthalmology, Pharmacology | Leave a comment

100 Cases in Orthopaedics and Rheumatology (I)

This book was decent, but it’s not as good as some of the books I’ve previously read in this series; in some of the books in the series the average length of the answer section is 2-3 pages, which is a format I quite like, whereas in this book the average is more like 1-2 pages – which is a bit too short in my opinion.

Below I have added some links related to the first half of the book’s coverage and a few observations from the book.

Acute haematogenous osteomyelitis. (“There are two principal types of acute osteomyelitis: •haematogenous osteomyelitis •direct or contiguous inoculation osteomyelitis. Acute haematogenous osteomyelitis is characterized by an acute infection of the bone caused by the seeding of the bacteria within the bone from a remote source. This condition occurs primarily in children. […] In general, osteomyelitis has a bimodal age distribution. Acute haematogenous osteomyelitis is primarily a disease in children. Direct trauma and contiguous focus osteomyelitis are more common among adults and adolescents than in children. Spinal osteomyelitis is more common in individuals older than 45 years.”)
Haemophilic arthropathy. (“Haemophilic arthropathy is a condition associated with clotting disorder leading to recurrent bleeding in the joints. Over time this can lead to joint destruction.”)
Avascular necrosis of the femoral head. Trendelenburg’s sign. Gaucher’s disease. Legg–Calvé–Perthes disease. Ficat and Arlet classification of avascular necrosis of femoral head.
Osteosarcoma. Codman triangle. Enneking Classification. (“A firm, irregular mass fixed to underlying structures is more suspicious of a malignant lesion.”)
Ewing’s sarcomaHaversian canal. (“This condition [ES] typically occurs in young patients and presents with pain and fever. [It] is the second most common primary malignant bone tumour (the first being osteosarcoma). The tumour is more common in males and affects children and young adults. The majority develop this between the ages of 10 and 20 years. […] The earliest symptom is pain, which is initially intermittent but becomes intense. Rarely, a patient may present with a pathological fracture. Eighty-five per cent of patients have chromosomal translocations associated with the 11/22 chromosome. Ewing’s sarcoma is potentially the most aggressive form of the primary bone tumours. […] Patients are usually assigned to one of two groups, the tumour being classified as either localized or metastatic disease. Tumours in the pelvis typically present late and are therefore larger with a poorer prognosis. Treatment comprises chemotherapy, surgical resection and/or radiotherapy. […] With localized disease, wide surgical excision of the tumour is preferred over radiotherapy if the involved bone is expendable (e.g. fibular, rib), or if radiotherapy would damage the growth plate. […] Non-metastatic disease survival rates are 55–70 per cent, compared to 22–33 per cent for metastatic disease. Patients require careful follow-up owing to the risk of developing osteosarcoma following radiotherapy, particularly in children in whom it can occur in up to 20 per cent of cases.”
Clavicle Fracture. Floating Shoulder.
Proximal humerus fractures.
Lateral condyle fracture of the humerus. Salter-Harris fracture. (“Humeral condyle fractures occur most commonly between 6 and 10 years of age. […] fractures often appear subtle on radiographs. […] Operative management is essential for all displaced fractures“).
Distal radius fracture. (“Colles’ fractures account for over 90 per cent of distal radius fractures. Any injury to the median nerve can produce paraesthesia in the thumb, index finger, and middle and radial border of the ring finger […]. There is a bimodal age distribution of fractures to the distal radius with two peaks occurring. The first peak occurs in people aged 18–25 years, and a second peak in older people (>65 years). High-energy injuries are more common in the younger group and low-energy injuries in the older group. Osteoporosis may play a role in the occurrence of this later fracture. In the group of patients between 60 and 69 years, women far outnumber men. […] Assessment with plain radiographs is all that is needed for most fractures. […] The majority of distal radius fractures can be treated conservatively.”)
Gamekeeper’s thumb. Stener lesion.
Subtrochanteric Hip Fracture.
Supracondylar Femur Fractures. (“There is a bimodal distribution of fractures based on age and gender. Most high-energy distal femur fractures occur in males aged between 15 and 50 years, while most low-energy fractures occur in osteoporotic women aged 50 or above. The most common high-energy mechanism of injury is a road traffic accident (RTA), and the most common low-energy mechanism is a fall. […] In general, […] non-operative treatment does not work well for displaced fractures. […] Operative intervention is also indicated in the presence of open fractures and injuries associated with vascular injury. […] Total knee replacement is effective in elderly patients with articular fractures and significant osteoporosis, or pre-existing arthritis that is not amenable to open reduction and internal fixation. Low-demand elderly patients with non- or minimally displaced fractures can be managed conservatively. […] In general, this fracture can take a minimum of 3-4 months to unite.”)
Supracondylar humerus fracture. Gartland Classification of Supracondylar Humerus Fractures. (“Prior to the treatment of supracondylar fractures, it is essential to identify the type. Examination of the degree of swelling and deformity as well as a neurological and vascular status assessment of the forearm is essential. A vascular injury may present with signs of an acute compartment syndrome with pain, paraesthesia, pallor, and pulseless and tight forearm. Injury to the brachial artery may present with loss of the distal pulse. However, in the presence of a weak distal pulse, major vessel injury may still be present owing to the collateral circulation. […] Vascular insult can lead to Volkmann ischaemic contracture of the forearm. […] Malunion of the fracture may lead to cubitus varus deformity.”)
Femoral Shaft Fractures.
Femoral Neck Fractures. Garden’s classification. (“Hip fractures are the most common reason for admission to an orthopaedic ward, usually caused by a fall by an elderly person. The average age of a person with a hip fracture is 77 years. Mortality is high: about 10 per cent of people with a hip fracture die within 1 month, and about one-third within 12 months. However, fewer than half of deaths are attributable to the fracture, reflecting the high prevalence of comorbidity. The mental status of the patient is also important: senility is associated with a three-fold increased risk of sepsis and dislocation of prosthetic replacement when compared with mentally alert patients. The one-year mortality rate in these patients is considerable, being reported as high as 50 per cent.”)
Tibia Shaft Fractures. (“The tibia is the most frequent site of a long-bone fracture in the body. […] Open fractures are surgical emergencies […] Most closed tibial fractures can be treated conservatively using plaster of Paris.”)
Tibial plateau fracture. Schatzker classification.
Compartment syndrome. (“This condition is an orthopaedic emergency and can be limb- and life-threatening. Compartment syndrome occurs when perfusion pressure falls below tissue pressure in a closed fascial compartment and results in microvascular compromise. At this point, blood flow through the capillaries stops. In the absence of flow, oxygen delivery stops. Hypoxic injury causes cells to release vasoactive substances (e.g. histamine, serotonin), which increase endothelial permeability. Capillaries allow continued fluid loss, which increases tissue pressure and advances injury. Nerve conduction slows, tissue pH falls due to anaerobic metabolism, surrounding tissue suffers further damage, and muscle tissue suffers necrosis, releasing myoglobin. In untreated cases the syndrome can lead to permanent functional impairment, renal failure secondary to rhabdomyolysis, and death. Patients at risk of compartment syndrome include those with high-velocity injuries, long-bone fractures, high-energy trauma, penetrating injuries such as gunshot wounds and stabbing, and crush injuries, as well as patients on anticoagulants with trauma. The patient usually complains of severe pain that is out of proportion to the injury. An assessment of the affected limb may reveal swelling which feels tense, or hard compartments. Pain on passive range of movement of fingers or toes of the affected limb is a typical feature. Late signs comprise pallor, paralysis, paraesthesia and a pulseless limb. Sensory nerves begin to lose conductive ability, followed by motor nerves. […] Fasciotomy is the definitive treatment for compartment syndrome. The purpose of fasciotomy is to achieve prompt and adequate decompression so as to restore the tissue perfusion.”)
Talus fracture. Hawkins sign. Avascular necrosis.
Calcaneal fracture. (“The most common situation leading to calcaneal fracture is a young adult who falls from a height and lands on his or her feet. […] Patients often sustain occult injuries to their lumbar or cervical spine, and the proximal femur. A thorough clinical and radiological investigation of the spine area is mandatory in patients with calcaneal fracture.”)
Idiopathic scoliosis. Adam’s forward bend test. Romberg test. Cobb angle.
Cauda equina syndrome. (“[Cauda equina syndrome] is an orthopaedic emergency. The condition is characterized by the red-flag signs comprising low back pain, unilateral or bilateral sciatica, saddle anaesthesia with sacral sparing, and bladder and bowel dysfunctions. Urinary retention is the most consistent finding. […] Urgent spinal orthopaedic or neurosurgical consulation is essential, with transfer to a unit capable of undertaking any definitive surgery considered necessary. In the long term, residual weakness, incontinence, impotence and/or sensory abnormalities are potential problems if therapy is delayed. […] The prognosis improves if a definitive cause is identified and appropriate surgical spinal decompression occurs early. Late surgical compression produces varying results and is often associated with a poorer outcome.”)
Developmental dysplasia of the hip.
OsteoarthritisArthroplasty. OsteotomyArthrodesis. (“Early-morning stiffness that gradually diminishes with activity is typical of osteoarthritis. […] Patients with hip pathology can sometimes present with knee pain without any groin or thigh symptoms. […] Osteoarthritis most commonly affects middle-aged and elderly patients. Any synovial joint can develop osteoarthritis. This condition can lead to degeneration of articular cartilage and is often associated with stiffness.”)
Prepatellar bursitis.
Baker’s cyst.
Meniscus tear. McMurray test. Apley’s test. Lachman test.
Anterior cruciate ligament injury.
Achilles tendon rupture. Thompson Test.
Congenital Talipes EquinovarusPonseti method. Pirani score. (“Club foot is bilateral in about 50 per cent of cases and occurs in approximately 1 in 800 births.”)
Charcot–Marie–Tooth disease. Pes cavus. Claw toe deformity. Pes planus.
Hallux valgus. Hallux Rigidus.
Mallet toe deformity. Condylectomy. Syme amputation. (“Mallet toes are common in diabetics with peripheral neuropathy. […] Pain and/or a callosity is often the presenting complaint. This may also lead to nail deformity on the toe. Most commonly the deformity is present in the second toe. […] Footwear modification […] should be tried first […] Surgical management of mallet toe is indicated if the deformity becomes painful.”)
Hammer Toe.
Lisfranc injury. Fleck sign. (“The Lisfranc joint, which represents the articulation between the midfoot and forefoot, is composed of the five TMT [tarsometatarsal] joints. […] A Lisfranc injury encompasses everything from a sprain to a complete disruption of normal anatomy through the TMT joints. […] Lisfranc injuries are commonly undiagnosed and carry a high risk of chronic secondary disability.”)
Charcot joint. (“Charcot arthropathy results in progressive destruction of bone and soft tissues at weight-bearing joints. In its most severe form it may cause significant disruption of the bony architecture, including joint dislocations and fractures. Charcot arthropathy can occur at any joint but most commonly affects the lower regions: the foot and ankle. Bilateral disease occurs in fewer than 10 per cent of patients. Any condition that leads to a sensory or autonomic neuropathy can cause a Charcot joint. Charcot arthropathy can occur as a complication of diabetes, syphilis, alcoholism, leprosy, meningomyleocele, spinal cord injury, syringomyelia, renal dialysis and congenital insensitivity to pain. In the majority of cases, non-operative methods are preferred. The principles of management are to provide immobilization of the affected joint and reduce any areas of stress on the skin. Immobilization is usually accomplished by casting.”)
Lateral epicondylitis (tennis elbow). (“For work-related lateral epicondylitis, a systematic review identified three risk factors: handling tools heavier than 1 kg, handling loads heavier than 20 kg at least ten times per day, and repetitive movements for more than two hours per day. […] Up to 95 per cent of patients with tennis elbow respond to conservative measures.”)
Medial Epicondylitis.
De Quervain’s tenosynovitis. Finkelstein test. Intersection syndrome. Wartenberg’s syndrome.
Trigger finger.
Adhesive capsulitis (‘frozen shoulder’). (“Frozen shoulder typically has three phases: the painful phase, the stiffening phase and the thawing phase. During the initial phase there is a gradual onset of diffuse shoulder pain lasting from weeks to months. The stiffening phase is characterized by a progressive loss of motion that may last up to a year. The majority of patients lose glenohumeral external rotation, internal rotation and abduction during this phase. The final, thawing phase ranges from weeks to months and constitutes a period of gradual motion improvement. Once in this phase, the patient may require up to 9 months to regain a fully functional range of motion. There is a higher incidence of frozen shoulder in patients with diabetes compared with the general population. The incidence among patients with insulin-dependent diabetes is even higher, with an increased frequency of bilateral frozen shoulder. Adhesive capsulitis has also been reported in patients with hyperthyroidism, ischaemic heart disease, and cervical spondylosis. Non-steroidal anti-inflammatory drugs (NSAIDs) are recommended in the initial treatment phase. […] A subgroup of patients with frozen shoulder syndrome often fail to improve despite conservative measures. In these cases, interventions such as manipulation, distension arthrography or open surgical release may be beneficial.” [A while back I covered some papers on adhesive capsulitis and diabetes here (part iii) – US].
Dupuytren’s Disease. (“Dupuytren’s contracture is a benign, slowly progressive fibroproliferative disease of the palmar fascia. […] The disease presents most commonly in the ring and little fingers and is bilateral in 45 per cent of cases. […] Dupuytren’s disease is more common in males and people of northern European origin. It can be associated with prior hand trauma, alcoholic cirrhosis, epilepsy (due to medications such as phenytoin), and diabetes. [“Dupuytren’s disease […] may be observed in up to 42% of adults with diabetes mellitus, typically in patients with long-standing T1D” – I usually don’t like such unspecific reported prevalences (what does ‘up to’ really mean?), but the point is that this is not a 1 in a 100 complication among diabetics; it seems to be a relatively common complication in type 1 DM – US] The prevalence increases with age. Mild cases may not need any treatment. Surgery is indicated in progressive contractures and established deformity […] Recurrence or extension of the disease after operation is not uncommon”).

July 1, 2018 Posted by | Books, Cancer/oncology, Diabetes, Medicine, Neurology | Leave a comment

Gastrointestinal complications of diabetes (I)

I really liked this book. It covered a lot of stuff also covered in Horowitz & Samsom’s excellent book on these topics, but it’s shorter and so probably easier for the relevant target group to justify reading. I recommend the book if you want to know more about these topics but don’t quite feel like reading a long textbook on these topics.

Below I’ve added some observations from the first half of the book. In the quotes below I’ve added some links and highlighted some key observations by the use of bold text.

Gastrointestinal (GI) symptoms occur more commonly in patients with diabetes than in the general population [2]. […] GI symptoms such as nausea, abdominal pain, bloating, diarrhea, constipation, and delayed gastric emptying occur in almost 75 % of patients with diabetes [3]. A majority of patients with GI symptoms stay undiagnosed or undertreated due to a lack of awareness of these complications among clinicians. […] Diabetes can affect the entire GI tract from the oral cavity and esophagus to the large bowel and anorectal region, either in isolation or in a combination. The extent and the severity of the presenting symptoms may vary widely depending upon which part of the GI tract is involved. In patients with long-term type 1 DM, upper GI symptoms seem to be particularly common [4]. Of the different types […] gastroparesis seems to be the most well known and most serious complication, occurring in about 50 % of patients with diabetes-related GI complications [5].”

The enteric nervous system (ENS) is an independent network of neurons and glial cells that spread from the esophagus up to the internal anal sphincter. […] the ENS regulates GI tract functions including motility, secretion, and participation in immune regulation [12, 13]. GI complications and their symptoms in patients with diabetes arise secondary to both abnormalities of gastric function (sensory and motor modality), as well as impairment of GI hormonal secretion [14], but these abnormalities are complex and incompletely understood. […] It has been known for a long time that diabetic autonomic neuropathy […] leads to abnormalities in the GI motility, sensation, secretion, and absorption, serving as the main pathogenic mechanism underlying GI complications. Recently, evidence has emerged to suggest that other processes might also play a role. Loss of the pacemaker interstitial cells of Cajal, impairment of the inhibitory nitric oxide-containing nerves, abnormal myenteric neurotransmission, smooth muscle dysfunction, and imbalances in the number of excitatory and inhibitory enteric neurons can drastically alter complex motor functions causing dysfunction of the enteric system [7, 11, 15, 16]. This dysfunction can further lead to the development of dysphagia and reflux esophagitis in the esophagus, gastroparesis, and dyspepsia in the stomach, pseudo-obstruction of the small intestine, and constipation, diarrhea, and incontinence in the colon. […] Compromised intestinal vascular flow arising due to ischemia and hypoxia from microvascular disease of the GI tract can also cause abdominal pain, bleeding, and mucosal dysfunction. Mitochondrial dysfunction has been implicated in the pathogenesis of gastric neuropathy. […] Another possible association between DM and the gastrointestinal tract can be infrequent autoimmune diseases associated with type I DM like autoimmune chronic pancreatitis, celiac disease (2–11 %), and autoimmune gastropathy (2 % prevalence in general population and three- to fivefold increase in patients with type 1 DM) [28, 29]. GI symptoms are often associated with the presence of other diabetic complications, especially autonomic and peripheral neuropathy [2, 30, 31]. In fact, patients with microvascular complications such as retinopathy, nephropathy, or neuropathy should be presumed to have GI abnormalities until proven otherwise. In a large cross-sectional questionnaire study of 1,101 subjects with DM, 57 % of patients reported at least one GI complication [31]. Poor glycemic control has also been found to be associated with increased severity of the upper GI symptoms. […] management of DM-induced GI complications is challenging, is generally suboptimal, and needs improvement.

Diabetes mellitus (DM) has multiple clinically important effects on the esophagus. Diabetes results in several esophageal motility disturbances, increases the risk of esophageal candidiasis, and increases the risk of Barrett’s esophagus and esophageal carcinoma. Finally, “black esophagus,” or acute esophageal necrosis, is also associated with DM. […] Esophageal dysmotility has been shown to be associated with diabetic neuropathy; however, symptomatic esophageal dysmotility is not often considered an important complication of diabetes. […] In general, the manometric effects of diabetes on the esophagus are not specific and mostly related to speed and strength of peristalsis. […] The pathological findings which amount to loss of cholinergic stimulation are consistent with the manometric findings in the esophagus, which are primarily related to slowed or weakened peristalsis. […] The association between DM and GERD is complex and conflicting. […] A recent meta-analysis suggests an overall positive association in Western countries [12]. […] The underlying pathogenesis of DM contributing to GERD is not fully elucidated, but is likely related to reduced acid clearance due to slow, weakened esophageal peristalsis. The association between DM and gastroesophageal reflux (GER) is well established, but the link between DM and GERD, which requires symptoms or esophagitis, is more complex because sensation may be blunted in diabetics with neuropathy. Asymptomatic gastroesophageal reflux (GER) confirmed by pH studies is significantly more frequent in diabetic patients than in healthy controls [13]. […] long-standing diabetics with neuropathy are at higher risk for GERD even if they have no symptoms. […] Abnormal pH and motility studies do not correlate very well with the GI symptoms of diabetics, possibly due to DM-related sensory dysfunction.”

Gastroparesis is defined as a chronic disorder characterized by delayed emptying of the stomach occurring in the absence of mechanical obstruction. It is a well-known and potentially serious complication of diabetes. […] Diabetic gastroparesis affects up to 40 % of patients with type 1 diabetes and up to 30 % of patients with type 2 diabetes [1, 2]. Diabetic gastroparesis generally affects patients with longstanding diabetes mellitus, and patients often have other diabetic complications […] For reasons that remain unclear, approximately 80 % of patients with gastroparesis are women [3]. […] In diabetes, delayed gastric emptying can often be asymptomatic. Therefore, the term gastroparesis should only be reserved for patients that have both delayed gastric emptying and upper gastrointestinal symptoms. Additionally, discordance between the pattern and type of symptoms and the magnitude of delayed gastric emptying is a well-established phenomenon. Accelerating gastric emptying may not improve symptoms, and patients can have symptomatic improvement while gastric emptying time remains unchanged. Furthermore, patients with severe symptoms can have mild delays in gastric emptying. Clinical features of gastroparesis include nausea, vomiting, bloating, abdominal pain, and malnutrition. […] Gastroparesis affects oral drug absorption and can cause hyperglycemia that is challenging to manage, in addition to unexplained hypoglycemia. […] Nutritional and caloric deficits are common in patients with gastroparesis […] Possible complications of gastroparesis include volume depletion with renal failure, malnutrition, electrolyte abnormalities, esophagitis, Mallory–Weiss tear (from vomiting), or bezoar formation. […] Unfortunately, there is a dearth of medications available to treat gastroparesis. Additionally, many of the medications used are based on older trials with small sample sizes […and some of them have really unpleasant side effects – US]. […] Gastroparesis can be associated with abdominal pain in as many as 50 % of patients with gastroparesis at tertiary care centers. There are no trials to guide the choice of agents. […] Abdominal pain […] is often difficult to treat [3]. […] In a subset of patients with diabetes [less than 10%, according to Horowitz & Samsom – US], gastric emptying can be abnormally accelerated […]. Symptoms are often difficult to distinguish from those with delayed gastric emptying. […] Worsening symptoms with a prokinetic agent can be a sign of possible accelerated emptying.”

“Diabetic enteropathy encompasses small intestinal and colorectal dysfunctions such as diarrhea, constipation, and/or fecal incontinence. It is more commonly seen in patients with long-standing diabetes, especially in those with gastroparesis. Development of diabetic enteropathy is complex and multifactorial. […] gastrointestinal symptoms and complications do not always correlate with the duration of diabetes, glycemic control, or with the presence of autonomic neuropathy, which is often assumed to be the major cause of many gastrointestinal symptoms. Other pathophysiologic processes operative in diabetic enteropathy include enteric myopathy and neuropathy; however, causes of these abnormalities are unknown [1]. […] Collectively, the effects of diabetes on several targets cause aberrations in gastrointestinal function and regulation. Loss of ICC, autonomic neuropathy, and imbalances in the number of excitatory and inhibitory enteric neurons can drastically alter complex motor functions such as peristalsis, reflexive relaxation, sphincter tone, vascular flow, and intestinal segmentation [5]. […] Diarrhea is a common complaint in DM. […] Etiologies of diarrhea in diabetes are multifactorial and include rapid intestinal transit, drug-induced diarrhea, small-intestine bacterial overgrowth, celiac disease, pancreatic exocrine insufficiency, dietary factors, anorectal dysfunction, fecal incontinence, and microscopic colitis [1]. […] It is important to differentiate whether diarrhea is caused by rapid intestinal transit vs. SIBO. […] This differentiation has key clinical implications with regard to the use of antimotility agents or antibiotics in a particular case. […] Constipation is a common problem seen with long-standing DM. It is more common than in general population, where the incidence varies from 2 % to 30 % [30]. It affects 60 % of the patients with DM and is more common than diarrhea [14]. […] There are no specific treatments for diabetes-associated constipation […] In most cases, patients are treated in the same way as those with idiopathic chronic constipation. […] Colorectal cancer is the third most common cancer in men and the second in women [33]. Individuals with type 2 DM have an increased risk of colorectal cancer when compared with their nondiabetic counterparts […] According to a recent large observational population-based cohort study, type 2 DM was associated with a 1.3-fold increased risk of colorectal cancer compared to the general population.”

Nonalcoholic fatty liver disease (NAFLD) is the main hepatic complication of obesity, insulin resistance, and diabetes and soon to become the leading cause for end-stage liver disease in the United States [1]. […] NAFLD is a spectrum of disease that ranges from steatosis (hepatic fat without significant hepatocellular injury) to nonalcoholic steatohepatitis (NASH; hepatic fat with hepatocellular injury) to advanced fibrosis and cirrhosis. As a direct consequence of the obesity epidemic, NAFLD is the most common cause of chronic liver disease, while NASH is the second leading indication for liver transplantation [1]. NAFLD prevalence is estimated at 25 % globally [2] and up to 30 % in the United States [3–5]. Roughly 30 % of individuals with NAFLD also have NASH, the progressive subtype of NAFLD. […] NASH is estimated at 22 % among patients with diabetes, compared to 5 % of the general population [4, 14]. […] Insulin resistance is strongly associated with NASH. […] Simple steatosis (also known as nonalcoholic fatty liver) is characterized by the presence of steatosis without ballooned hepatocytes (which represents hepatocyte injury) or fibrosis. Mild inflammation may be present. Simple steatosis is associated with a very low risk of progressive liver disease and liver-related mortality. […] Patients with NASH are at risk for progressive liver fibrosis and liver-related mortality, cardiovascular complications, and hepatocellular carcinoma (HCC) even in the absence of cirrhosis [26]. Liver fibrosis stage progresses at an estimated rate of one stage every 7 years [27]. Twenty percent of patients with NASH will eventually develop liver cirrhosis [9]. […] The risk of cardiovascular disease is increased across the entire NAFLD spectrum. […] Cardiovascular risk reduction should be aggressively managed in all patients.

 

June 17, 2018 Posted by | Books, Cancer/oncology, Cardiology, Diabetes, Gastroenterology, Medicine, Neurology | Leave a comment

Alcohol and Aging (II)

I gave the book 3 stars on goodreads.

As is usual for publications of this nature, the book includes many chapters that cover similar topics and so the coverage can get a bit repetitive if you’re reading it from cover to cover the way I did; most of the various chapter authors obviously didn’t read the other contributions included in the book, and as each chapter is meant to stand on its own you end up with a lot of chapter introductions which cover very similar topics. If you can disregard such aspects it’s a decent book, which covers a wide variety of topics.

Below I have added some observations from some of the chapters of the book which I did not cover in my first post.

It is widely accepted that consuming heavy amounts of alcohol and binge drinking are detrimental to the brain. Animal studies that have examined the anatomical changes that occur to the brain as a consequence of consuming alcohol indicate that heavy alcohol consumption and binge drinking leads to the death of existing neurons [10, 11] and prevents production of new neurons [12, 13]. […] While animal studies indicate that consuming even moderate amounts of alcohol is detrimental to the brain, the evidence from epidemiological studies is less clear. […] Epidemiological studies that have examined the relationship between late life alcohol consumption and cognition have frequently reported that older adults who consume light to moderate amounts of alcohol are less likely to develop dementia and have higher cognitive functioning compared to older adults who do not consume alcohol. […] In a meta-analysis of 15 prospective cohort studies, consuming light to moderate amounts of alcohol was associated with significantly lower relative risk (RR) for Alzheimer’s disease (RR=0.72, 95% CI=0.61–0.86), vascular dementia (RR=0.75, 95% CI=0.57–0.98), and any type of dementia (RR=0.74, 95% CI=0.61–0.91), but not cognitive decline (RR=0.28, 95 % CI=0.03–2.83) [31]. These findings are consistent with a previous meta-analysis by Peters et al. [33] in which light to moderate alcohol consumption was associated with a decreased risk for dementia (RR=0.63, 95 % CI=0.53–0.75) and Alzheimer’s disease (RR=0.57, 95 % CI=0.44–0.74), but not vascular dementia (RR=0.82, 95% CI=0.50–1.35) or cognitive decline RR=0.89, 95% CI=0.67–1.17). […] Mild cognitive impairment (MCI) has been used to describe the prodromal stage of Alzheimer’s disease […]. There is no strong evidence to suggest that consuming alcohol is protective against MCI [39, 40] and several studies have reported non-significant findings [41–43].”

The majority of research on the relationship between alcohol consumption and cognitive outcomes has focused on the amount of alcohol consumed during old age, but there is a growing body of research that has examined the relationship between alcohol consumption during middle age and cognitive outcomes several years or decades later. The evidence from this area of research is mixed with some studies not detecting a significant relationship [17, 58, 59], while others have reported that light to moderate alcohol consumption is associated with preserved cognition [60] and decreased risk for cognitive impairment [31, 61, 62]. […] Several epidemiological studies have reported that light to moderate alcohol consumption is associated with a decreased risk for stroke, diabetes, and heart disease [36, 84, 85]. Similar to the U-shaped relationship between alcohol consumption and dementia, heavy alcohol consumption has been associated with poor health [86, 87]. The decreased risk for several metabolic and vascular health conditions for alcohol consumers has been attributed to antioxidants [54], greater concentrations of high-density lipoprotein cholesterol in the bloodstream [88], and reduced blood clot formation [89]. Stroke, diabetes, heart disease, and related conditions have all been associated with lower cognitive functioning during old age [90, 91]. The reduced prevalence of metabolic and vascular health conditions among light to moderate alcohol consumers may contribute to the decreased risk for dementia and cognitive decline for older adults who consume alcohol. A limitation of the hypothesis that the reduced risk for dementia among light and moderate alcohol consumers is conferred through the reduced prevalence of adverse health conditions associated with dementia is the possibility that this relationship is confounded by reverse causality. Alcohol consumption decreases with advancing age and adults may reduce their alcohol consumption in response to the onset of adverse health conditions […] the higher prevalence of dementia and lower cognitive functioning among abstainers may be due in part to their worse health rather than their alcohol consumption.”

A limitation of large cohort studies is that subjects who choose not to participate or are unable to participate are often less healthy than those who do participate. Non-response bias becomes more pronounced with age because only subjects who have survived to old age and are healthy enough to participate are observed. Studies on alcohol consumption and cognition are sensitive to non-response bias because light and moderate drinkers who are not healthy enough to participate in the study will not be observed. Adults who survive to old age despite consuming very high amounts of alcohol represent an even more select segment of the general population because they may have genetic, behavioral, health, social, or other factors that protect them against the negative effects of heavy alcohol consumption. As a result, the analytic sample of epidemiological studies is more likely to be comprised of “healthy” drinkers, which biases results in favor of finding a positive effect of light to moderate alcohol consumption for cognition and health in general. […] The incidence of Alzheimer’s disease doubles every 5 years after 65 years of age [94] and nearly 40% of older adults aged 85 and over are diagnosed with Alzheimer’s disease [7]. The relatively old age of onset for most dementia cases means the observed protective effect of light to moderate alcohol consumption for dementia may be due to alcohol consumers being more likely to die or drop out of a study as a result of their alcohol consumption before they develop dementia. This bias may be especially strong for heavy alcohol consumers. Not properly accounting for death as a competing outcome has been observed to artificially increase the risk of dementia among older adults with diabetes [95] and the effect that death and other competing outcomes may have on the relationship between alcohol consumption and dementia risk is unclear. […] The majority of epidemiological studies that have studied the relationship between alcohol consumption and cognition treat abstainers as the reference category. This can be problematic because often times the abstainer or non-drinking category includes older adults who stopped consuming alcohol because of poor health […] Not differentiating former alcohol consumers from lifelong abstainers has been found to explain some but not all of the benefit of alcohol consumption for preventing mortality from cardiovascular causes [96].”

“It is common for people to engage in other behaviors while consuming alcohol. This complicates the relationship between alcohol consumption and cognition because many of the behaviors associated with alcohol consumption are positively and negatively associated with cognitive functioning. For example, alcohol consumers are more likely to smoke than non-drinkers [104] and smoking has been associated with an increased risk for dementia and cognitive decline [105]. […] The relationship between alcohol consumption and cognition may also differ between people with or without a history of mental illness. Depression reduces the volume of the hippocampus [106] and there is growing evidence that depression plays an important role in dementia. Depression during middle age is recognized as a risk factor for dementia [107], and high depressive symptoms during old age may be an early symptom of dementia [108]. Middle aged adults with depression or other mental illness who self-medicate with alcohol may be at especially high risk for dementia later in life because of synergistic effects that alcohol and depression has on the brain. […] While current evidence from epidemiological studies indicates that consuming light to moderate amounts of alcohol, in particular wine, does not negatively affect cognition and in many cases is associated with cognitive health, adults who do not consume alcohol should not be encouraged to increase their alcohol consumption until further research clarifies these relationships. Inconsistencies between studies on how alcohol consumption categories are defined make it difficult to determine the “optimal” amount of alcohol consumption to prevent dementia. It is likely that the optimal amount of alcohol varies according to a person’s gender, as well as genetic, physiological, behavioral, and health characteristics, making the issue extremely complex.”

Falls are the leading cause of both fatal and nonfatal injuries among older adults, with one in three older adults falling each year, and 20–30% of people who fall suffer moderate to severe injuries such as lacerations, hip fractures, and head traumas. In fact, falls are the foremost cause of both fractures and traumatic brain injury (TBI) among older adults […] In 2013, 2.5 million nonfatal falls among older adults were treated in ED and more than 734,000 of these patients were hospitalized. […] Our analysis of the 2012 Nationwide Emergency Department Sample (NEDS) data set show that fall-related injury was a presenting problem among 12% of all ED visits by those aged 65+, with significant differences among age groups: 9% among the 65–74 age group, 12 % among the 75–84 age group, and 18 % among the 85+ age group [4]. […] heavy alcohol use predicts fractures. For example, among those 55+ years old in a health survey in England, men who consumed more than 8 units of alcohol and women who consumed more than 6 units on their heaviest drinking day in the past week had significantly increased odds of fractures (OR =1.65, 95% CI =1.37–1.98 for men and OR=2.07, 95% CI =1.28–3.35 for women) [63]. […] The 2008–2009 Canadian Community Health Survey-Healthy Aging also showed that consumption of at least one alcoholic drink per week increased the odds of falling by 40 % among those 65+ years [57].”

I at first was not much impressed by the effect sizes mentioned above because there are surely 100 relevant variables they didn’t account for/couldn’t account for, but then I thought a bit more about it. An important observation here – they don’t mention it in the coverage, but it sprang to mind – is that if sick or frail elderly people consume less alcohol than their more healthy counterparts, and are more likely to not consume alcohol (which they do, and which they are, we know this), and if frail or sick(er) elderly people are more likely to suffer a fall/fracture than are people who are relatively healthy (they are, again, we know this), well, then you’d expect consumption of alcohol to be found to have a ‘protective effect’ simply due to confounding by (reverse) indication (unless the researchers were really careful about adjusting for such things, but no such adjustments are mentioned in the coverage, which makes sense as these are just raw numbers being reported). The point is that the null here should not be that ‘these groups should be expected to have the same fall rate/fracture rate’, but rather ‘people who drink alcohol should be expected to be doing better, all else equal’ – but they aren’t, quite the reverse. So ‘the true effect size’ here may be larger than what you’d think.

I’m reasonably sure things are a lot more complicated than the above makes it appear (because of those 100 relevant variables we were talking about…), but I find it interesting anyway. Two more things to note: 1. Have another look at the numbers above if they didn’t sink in the first time. This is more than 10% of emergency department visits for that age group. Falls are a really big deal. 2. Fractures in the elderly are also a potentially really big deal. Here’s a sample quote: “One-fifth of hip fracture victims will die within 6 months of the injury, and only 50% will return to their previous level of independence.” (link). In some contexts, a fall is worse news than a cancer diagnosis, and they are very common events in the elderly. This also means that even relatively small effect sizes here can translate into quite large public health effects, because baseline incidence is so high.

The older adult population is a disproportionate consumer of prescription and over-the-counter medications. In a nationally representative sample of community-dwelling adults aged 57–84 years from the National Social Life, Health, and Aging Project (NSHAP) in 2005–2006, 81 % regularly used at least one prescription medication on a regular basis and 29% used at least five prescription medications. Forty-two percent used at least one nonprescription medication and concurrent use with a prescription medication was common, with 46% of prescription medication users also using OTC medications [2]. Prescription drug use by older adults in the U.S. is also growing. The percentage of older adults taking at least one prescription drug in the last 30 days increased from 73.6% in 1988–1994 to 89.7 % in 2007–2010 and the percentage taking five or more prescription drugs in the last 30 days increased from 13.8% in 1988–1994 to 39.7 % in 2007–2010 [3].”

The aging process can affect the response to a medication by altering its pharmacokinetics and pharmacodynamics [9, 10]. Reduced gastrointestinal motility and gastric acidity can alter the rate or extent of drug absorption. Changes in body composition, including decreased total body water and increased body fat can alter drug distribution. For alcohol, changes in body composition result in higher blood alcohol levels in older adults compared to younger adults after the same dose or quantity  of alcohol consumed. Decreased size of the liver, hepatic blood flow, and function of Phase I (oxidation, reduction, and hydrolysis) metabolic pathways result in reduced drug metabolism and increased drug exposure for drugs that undergo Phase I metabolism. Phase II hepatic metabolic pathways are generally preserved with aging. Decreased size of the kidney, renal blood flow, and glomerular filtration result in slower elimination of medications and metabolites by the kidney and increased drug exposure for medications that undergo renal elimination. Age-related impairment of homeostatic mechanisms and changes in receptor number and function can result in changes in pharmacodynamics as well. Older adults are generally more sensitive to the effects of medications and alcohol which act on the central nervous system for example. The consequences of these physiologic changes with aging are that older adults often experience increased drug exposure for the same dose (higher drug concentrations over time) and increased sensitivity to medications (greater response at a given drug concentration) than their younger counterparts.”

“Aging-related changes in physiology are not the only sources of variability in pharmacokinetics and pharmacodynamics that must be considered for an individual person. Older adults experience more chronic diseases that may decrease drug metabolism and renal elimination than younger cohorts. Frailty may result in further decline in drug metabolism, including Phase II metabolic pathways in the liver […] Drug interactions must also be considered […] A drug interaction is defined as a clinically meaningful change in the effect of one drug when coadministered with another drug [12]. Many drugs, including alcohol, have the potential for a drug interaction when administered concurrently, but whether a clinically meaningful change in effect occurs for a specific person depends on patient-specifc factors including age. Drug interactions are generally classified as pharmacokinetic interactions, where one drug alters the absorption, distribution, metabolism, or elimination of another drug resulting in increased or decreased drug exposure, or pharmacodynamic interactions, where one drug alters the response to another medication through additive or antagonistic pharmacologic effects [13]. An adverse drug event occurs when a pharmacokinetic or pharmacodynamic interaction or combination of both results in changes in drug exposure or response that lead to negative clinical outcomes. The adverse drug event could be a therapeutic failure if drug exposure is decreased or the pharmacologic response is antagonistic. The adverse drug event could be drug toxicity if the drug exposure is increased or the pharmacologic response is additive or synergistic. The threshold for experiencing an adverse event is often lower in older adults due to physiologic changes with aging and medical comorbidities, increasing their risk of experiencing an adverse drug event when medications are taken concurrently.”

“A large number of potential medication–alcohol interactions have been reported in the literature. Mechanisms of these interactions range from pharmacokinetic interactions affecting either alcohol or medication exposure to pharmacodynamics interactions resulting in exaggerated response. […] Epidemiologic evidence suggests that concurrent use of alcohol and medications among older adults is common. […] In a nationally representative U.S. sample of community-dwelling older adults in the National Social Life, Health and Aging Project (NSHAP) 2005–2006, 41% of participants reported consuming alcohol at least once per week and 20% were at risk for an alcohol–medication interaction because they were using both alcohol and alcohol-interacting medications on a regular basis [17]. […] Among participants in the Pennsylvania Assistance Contract for the Elderly program (aged 65–106 years) taking at least one prescription medication, 77% were taking an alcohol-interacting medication and 19% of the alcohol-interacting medication users reported concurrent use of alcohol [18]. […] Although these studies do not document adverse outcomes associated with alcohol–medication interactions, they do document that the potential exists for many older adults. […] High prevalence of concurrent use of alcohol and alcohol-interacting medications have also been reported in Australian men (43% of sedative or anxiolytic users were daily drinkers) [19], in older adults in Finland (42% of at-risk alcohol users were also taking alcohol-interacting medications) [20], and in older Irish adults (72% of participants were exposed to alcohol-interacting medications and 60% of these reported concurrent alcohol use) [21]. Drinking and medication use patterns in older adults may differ across countries, but alcohol–medication interactions appear to be a worldwide concern. […] Polypharmacy in general, and psychotropic burden specifically, has been associated with an increased risk of experiencing a geriatric syndrome such as falls or delirium, in older adults [26, 27]. Based on its pharmacology, alcohol can be considered as a psychotropic drug, and alcohol use should be assessed as part of the medication regimen evaluation to support efforts to prevent or manage geriatric syndromes. […] Combining alcohol and CNS active medications can be particularly problematic […] Older adults suffering from sleep problems or pain may be a particular risk for alcohol–medication interaction-related adverse events.”

In general, alcohol use in younger couples has been found to be highly concordant, that is, individuals in a relationship tend to engage in similar drinking behaviors [67,68]. Less is known, however, about alcohol use concordance between older couples. Graham and Braun [69] examined similarities in drinking behavior between spouses in a study of 826 community-dwelling older adults in Ontario, Canada. Results showed high concordance of drinking between spouses — whether they drank at all, how much they drank, and how frequently. […] Social learning theory suggests that alcohol use trajectories are strongly influenced by attitudes and behaviors of an individual’s social networks, particularly family and friends. When individuals engage in social activities with family and friends who approve of and engage in drinking, alcohol use, and misuse are reinforced [58, 59]. Evidence shows that among older adults, participation in social activities is correlated with higher levels of alcohol consumption [34, 60]. […] Brennan and Moos [29] […] found that older adults who reported less empathy and support from friends drank more alcohol, were more depressed, and were less self-confident. More stressors involving friends were associated with more drinking problems. Similar to the findings on marital conflict […], conflict in close friendships can prompt alcohol-use problems; conversely, these relationships can suffer as a result of alcohol-related problems. […] As opposed to social network theory […], social selection theory proposes that alcohol consumption changes an individual’s social context [33]. Studies among younger adults have shown that heavier drinkers chose partners and friends who approve of heavier drinking [70] and that excessive drinking can alienate social networks. The Moos study supports the idea that social selection also has a strong influence on drinking behavior among older adults.”

Traditionally, treatment studies in addiction have excluded patients over the age of 65. This bias has left a tremendous gap in knowledge regarding treatment outcomes and an understanding of the neurobiology of addiction in older adults.

Alcohol use causes well-established changes in sleep patterns, such as decreased sleep latency, decreased stage IV sleep, and precipitation or aggravation of sleep apnea [101]. There are also age-associated changes in sleep patterns including increased REM episodes, a decrease in REM length, a decrease in stage III and IV sleep, and increased awakenings. Age-associated changes in sleep can all be worsened by alcohol use and depression. Moeller and colleagues [102] demonstrated in younger subjects that alcohol and depression had additive effects upon sleep disturbances when they occurred together [102]. Wagman and colleagues [101] also have demonstrated that abstinent alcoholics did not sleep well because of insomnia, frequent awakenings, and REM fragmentation [101]; however, when these subjects ingested alcohol, sleep periodicity normalized and REM sleep was temporarily suppressed, suggesting that alcohol use could be used to self-medicate for sleep disturbances. A common anecdote from patients is that alcohol is used to help with sleep problems. […] The use of alcohol to self-medicate is considered maladaptive [34] and is associated with a host of negative outcomes. […] The use of alcohol to aid with sleep has been found to disrupt sleep architecture and cause sleep-related problems and daytime sleepiness [35, 36, 46]. Though alcohol is commonly used to aid with sleep initiation, it can worsen sleep-related breathing disorders and cause snoring and obstructive sleep apnea [36].”

Epidemiologic studies have clearly demonstrated that comorbidity between alcohol use and other psychiatric symptoms is common in younger age groups. Less is known about comorbidity between alcohol use and psychiatric illness in late life [88]. […] Blow et al. [90] reviewed the diagnosis of 3,986 VA patients between ages 60 and 69 presenting for alcohol treatment [90]. The most common comorbid psychiatric disorder was an affective disorder found in 21 % of the patients. […] Blazer et al. [91] studied 997 community dwelling elderly of whom only 4.5% had a history of alcohol use problems [91]; […] of these subjects, almost half had a comorbid diagnosis of depression or dysthymia. Comorbid depressive symptoms are not only common in late life but are also an important factor in the course and prognosis of psychiatric disorders. Depressed alcoholics have been shown to have a more complicated clinical course of depression with an increased risk of suicide and more social dysfunction than non-depressed alcoholics [9296]. […]  Alcohol use prior to late life has also been shown to influence treatment of late life depression. Cook and colleagues [94] found that a prior history of alcohol use problems predicted a more severe and chronic course for depression [94]. […] The effect of past heavy alcohol use is [also] highlighted in the findings from the Liverpool Longitudinal Study demonstrating a fivefold increase in psychiatric illness among elderly men who had a lifetime history of 5 or more years of heavy drinking [24]. The association between heavy alcohol consumption in earlier years and psychiatric morbidity in later life was not explained by current drinking habits. […] While Wernicke-Korsakoff’s syndrome is well described and often caused by alcohol use disorders, alcohol-related dementia may be difficult to differentiate from Alzheimer’s disease. Clinical diagnostic criteria for alcohol-related dementia (ARD) have been proposed and now validated in at least one trial, suggesting a method for distinguishing ARD, including Wernicke-Korsakoff’s syndrome, from other types of dementia [97, 98]. […] Finlayson et al. [100] found that 49 of 216 (23%) elderly patients presenting for alcohol treatment had dementia associated with alcohol use disorders [100].”

 

May 24, 2018 Posted by | Books, Demographics, Epidemiology, Medicine, Neurology, Pharmacology, Psychiatry, Statistics | Leave a comment

100 cases in emergency medicine and critical care (I)

“This book has been written for medical students, doctors and nurse practitioners. One of the best methods of learning is case-based learning. This book presents a hundred such ‘cases’ or ‘patients’ which have been arranged by system. Each case has been written to stand alone […] the focus of each case is to recognise the initial presentation, the underlying pathophysiology, and to understand broad treatment principles.”

I really liked the book; as was also the case for the surgery book I recently read the cases included in these publications are slightly longer than they were in some of the previous publications in the series I’ve read, and I think this makes a big difference in terms of how much you actually get out of each case.

Below I have added some links and quotes related to the first half of the book’s coverage.

Tracheostomy.
Malnutrition (“it is estimated that around a quarter of hospital inpatients are inadequately nourished. This may be due to increased nutritional requirements […], nutritional losses (e.g. malabsorption, vomiting, diarrhoea) or reduced intake […] A patient’s basal energy expenditure is doubled in head injuries and burns.”)
Acute Adult Supraglottitis. (“It is important to appreciate that halving the radius of the airway will increase its resistance by 16 times (Poiseuille’s equation), and hearing stridor means there is around 75% airway obstruction.”)
Out-of-hospital cardiac arrest. (“After successful resuscitation from an OHCA, only 10% of patients will survive to discharge, and many of these individuals will have significant neurologic disability.”)
Bacterial meningitis. (“Meningococcal meningitis has a high mortality, with 10%-15% of patients dying of the disease despite appropriate therapy.”)
Diabetic ketoacidosis.
Anaphylaxis (“Always think of anaphylaxis when seeing patients with skin/mucosal symptoms, respiratory difficulty and/or hypotension, especially after exposure to a potential allergen.”)
Early goal-directed therapy. (“While randomised evidence on the benefit of [this approach] is conflicting, it is standard practice in most centres.” I’m not sure I’d agree with the authors that the evidence is ‘conflicting’, it looks to me like it’s reasonably clear at this point: “In this meta-analysis of individual patient data, EGDT did not result in better outcomes than usual care and was associated with higher hospitalization costs across a broad range of patient and hospital characteristics.”)
Cardiac tamponade. Hypovolaemic shock. Permissive hypotensionFocused Assessment with Sonography in Trauma (FAST). (“Shock refers to inadequate tissue perfusion and tissue oxygenation. The commonest cause in an injured patient is hypovolaemic shock due to blood loss, but other causes include cardiogenic shock due to myocardial dysfunction, neurogenic shock due to sympathetic dysfunction or obstructive shock due to obstruction of the great vessels or heart. […] tachycardia, cool skin and reduced pulse pressure are early signs of shock until proven otherwise.”)
Intravenous therapy. A Comparison of Albumin and Saline for Fluid Resuscitation in the Intensive Care Unit.
Thermal burns. Curling’s ulcer. Escharotomy. Wallace rule of nines. Fluid management in major burn injuries. (“Alkali burns are more harmful than acidic. […] Electrical burns cause more destruction than the external burn may suggest. They are associated with internal destruction, as the path of least resistance is nerves and blood vessels. They can also cause arrhythmias and an electrocardiogram should be performed.”)
Steven Johnson syndrome. Nikolsky’s sign. SCORTEN scale.
Cardiac arrest. (“The mantra in the ED is that ‘you are not dead until you are warm and dead'”).
Myocardial infarction. (“The most important goal of the acute management of STEMI is coronary reperfusion, which may be achieved either by percutaneous coronary intervention (PCI) or use of fibrinolytic agents (thrombolysis). PCI is the preferred strategy if it can be delivered within 120 minutes of first medical contact (and ideally within 90 minutes) […] several randomised trials have shown that PCI provides improved short- and long-term survival outcomes compared to fibrinolysis, providing it can be performed within the appropriate time frame.”)
Asthma exacerbation. (“the prognosis for asthmatics admitted to the Intensive Care Unit is guarded, with an in-hospital mortality of 7% in those who are mechanically ventilated.”)
Acute exacerbation of COPD. Respiratory Failure.
Pulmonary embolism. CT pulmonary angiography. (“Obstructive cardiopulmonary disease is the main diagnosis to exclude in patients presenting with shortness of breath and syncope.”)
Sepsis. Sepsis Six. qSOFA. (“The main clinical features of sepsis include hypotension […], tachycardia […], a high (>38.3°C) or low (<36°C) temperature, altered mental status and signs of peripheral shutdown (cool skin, prolonged capillary refill, cyanosis) in severe cases. […] Sepsis is associated with substantial in-hospital morbidity and mortality, and an increased risk of death and re-admission to hospital even if the patient survives until discharge. Prognostic factors in sepsis include patient factors (increasing age, higher comorbidity), site of infection (urosepsis is associated with better outcomes compared to other sources), type of pathogen (nosocomial infections have higher mortality), early administration of antibiotics (which may reduce mortality by 50%) and restoration of perfusion.”)
Acute kidney injury. (“Classically there are three major causative categories of AKI: (i) pre-renal (i.e. hypoperfusion), (ii) renal (i.e. an intrinsic process with the kidneys) and (iii) post-renal (i.e. urinary tract obstruction). The initial evaluation should attempt to determine which of these are leading to AKI in the patient. […] two main complications that arise with AKI [are] volume and electrolyte issues.”)
Acute chest syndrome.
Thrombotic thrombocytopenic purpura. Schistocyte. Plasmapheresis.
Lower gastrointestinal bleeding. WarfarinProthrombin complex concentrate. (“Warfarin is associated with a 1%-3% risk of bleeding each year in patients with atrial fibrillation, and the main risk factors for this include presence of comorbities, interacting medications, poor patient compliance, acute illness and dietary variation in vitamin K intake.”)
Acute back pain. Malignant spinal cord compression (-MSCC). (“Acute back pain is not an uncommon reason for presentation to the Emergency Department […] Although the majority of such presentations represent benign pathology, it is important to exclude more serious pathology such as cord or cauda equina compression, infection or abscess. Features in the history warranting greater concern include a prior history of cancer, recent infection or steroid use, fever, pain in the thoracic region, pain that improves with rest and the presence of urinary symptoms. Similarly, ‘red flag’ examination findings include gait ataxia, generalized weakness, upper motor neurone signs (clonus, hyper-reflexia, extensor plantars), a palpable bladder, saddle anaesthesia and reduced anal tone. […] MSCC affects up to 5% of all cancer patients and is the first manifestation of cancer in a fifth of patients.”)
Neutropenic sepsis. (“Neutropaenic sepsis […] arises as a result of cytotoxic chemotherapy suppressing the bone marrow, leading to depletion of white blood cells and leaving the individual vulnerable to infection. It is one of the most common complications of cancer therapy, carrying a significant mortality rate of ~5%-10%, and should be regarded as a medical emergency. Any patient receiving chemotherapy and presenting with a fever should be assumed to have neutropaenic sepsis until proven otherwise.”)
Bacterial Pneumonia. CURB-65 Pneumonia Severity Score.
Peptic ulcer diseaseUpper gastrointestinal bleeding. Glasgow-Blatchford score. Rockall score.
Generalised tonic-clonic seizure. Status Epilepticus.
“Chest pain is an extremely common presentation in the ED […] Key features that may help point towards particular diagnoses include • Location and radiation – Central chest pain that radiates to the face, neck or arms is classic for MI, whereas the pain may be more posterior (between should blades) in aortic dissection and unilateral in lung disease. • Onset – Sudden or acute onset pain usually indicates a vascular cause (e.g. PE or aortic dissection), whereas cardiac chest pain is typically more subacute in onset and increases over time. • Character – Cardiac pain is usually described as crushing but may often be a gnawing discomfort, whereas pain associated with aortic dissection and gastrointestinal disorders is usually tearing/ripping and burning, respectively. • Exacerbation/alleviation […] myocardial ischaemia will manifest as pain brought on by exercise and relieved by rest, which is a good discriminator between cardiac and non-cardiac pain.”
Syncope. Mobitz type II AV block. (The differential diagnosis for syncope is seizure, and the two may be distinguished by the absence of a quick or spontaneous recovery with a seizure, where a post-ictal state (sleepiness, confusion, lethargy) is present.”)
Atrial Fibrillation. CHADSVASC and HASBLED risk scores. (“AF with rapid ventricular rates is generally managed with control of heart rates through use of beta-blockers or calcium-channel blockers. • Unstable patients with AF may require electrical cardioversion to restore sinus rhythm.”)
Typhoid fever. Dysentery.
Alcohol toxicity. (“Differentials which may mimic acute alcohol intoxication include • Hypoglycemia • Electrolyte disturbance • Vitamin depletion (B12/folate) • Head trauma • Sepsis • Other toxins or drug overdose • Other causes for CNS depression”)
Tricyclic Antidepressant Toxicity. (“Over 50% of suicidal overdoses involve more than one medication and are often taken with alcohol.”)
Suicide. SADPERSONS scale. (“Intentional self-harm results in around 150,000 attendances to the ED [presumably ‘every year’ – US]. These patients are 100 times more likely to commit suicide within the next year compared to the general population. Self-harm and suicide are often used interchangeably, but are in fact two separate entities. Suicide is a self-inflicted intentional act to cause death, whereas self-harm is a complex behaviour to inflict harm but not associated with the thought of dying – a method to relieve mental stress by inflicting physical pain.”)
Cauda equina syndrome (-CES). (“signs and symptoms of lower extremity weakness and pain developing acutely after heavy lifting should raise suspicion for a herniated intervertebral disc, which is the commonest cause of CES. […] CES is a neurosurgical emergency. The goal is to prevent irreversible loss of bowel and bladder function and motor function of the lower extremities. […] A multitude of alternative diagnoses may masquerade as CES – stroke, vascular claudication, deep venous thrombosis, muscle cramps and peripheral neuropathy.”)
Concussion.
Subarachnoid hemorrhage. Arteriovenous malformation.
Ischemic Stroke. AlteplaseMechanical thrombectomy for acute ischemic stroke. (“evaluation and treatment should be based on the understanding that the damage that is done (infarcted brain) is likely to be permanent, and the goal is to prevent further damage (ischaemic brain) and treat reversible causes (secondary prevention). Along those lines, time is critical to the outcome of the patient.”)
Mechanical back pain. Sciatica.
Dislocated shoulder. Bankart lesion. Hill-Sachs lesion. Kocher’s method.
Supracondylar Humerus Fractures. (“Supracondular fractures in the adult are relatively uncommon but are seen in major trauma or in elderly patients where bone quality may be compromised. Elbow fractures need careful neurovascular evaluation […] There are three major nerves that pass through the region: 1. The median nerve […] 2. The radial nerve […] 3. The ulnar nerve […] It is important to assess these three nerves and to document their function individually. The brachial artery passes through the cubital fossa and may be directly injured by bone fragments or suffer intimal damage. […] This is a true orthopaedic and vascular emergency as the upper limb can only tolerate an ischaemia time of around 90 minutes before irreparable damage is sustained.”)
Boxer’s fracture.

May 2, 2018 Posted by | Books, Cancer/oncology, Cardiology, Infectious disease, Medicine, Nephrology, Neurology, Psychiatry, Studies | Leave a comment

Endocrinology (part 6 – neuroendocrine disorders and Paget’s disease)

I’m always uncertain as to how much content to cover when covering books like this one, and I usually cover handbooks in less detail (relatively) than I cover other books because of the amount of work it takes to cover all topics of interest – however I didn’t feel after writing my last post in the series that I had really finished with this book, in terms of blogging it; in fact I remember distinctly feeling a bit annoyed towards the end of writing my fifth post by the fact that I didn’t find that I could justify covering the detailed account of Paget’s disease included in the last part of the chapter, even though all of that stuff was new knowledge to me, and quite interesting – but these posts take some effort, and sometimes I cut them short just to at least blog something, rather than just have an unpublished draft lying around.

In this post I’ll first include some belated coverage of Paget’s disease, which is from the book’s chapter 6, and then I’ll cover some of the stuff included in chapter 8 of the book, about neuroendocrine disorders. Chapter 8 deals exclusively with various types of (usually quite rare) tumours. I decided to not cover chapter 7, which is devoted to paediatric endocrinology.

“Paget’s disease is the result of greatly local bone turnover, which occurs particularly in the elderly […] The 1° abnormality in Paget’s disease is gross overactivity of the osteoclasts, resulting in greatly increased ↑ bone resorption. This secondarily results in ↑ osteoblastic activity. The new bone is laid down in a highly disorganized manner […] Paget’s disease can affect any bone in the skeleton […] In most patients, it affects several sites, but, in about 20% of cases, a single bone is affected (monostotic disease). Typically, the disease will start in one end of a long bone and spread along the bone at a rate of about 1cm per year. […] Paget’s disease alters the mechanical properties of the bone. Thus, pagetic bones are more likely to bend under normal physiological loads and are thus liable to fracture. […] Pagetic bones are also larger than their normal counterparts. This can lead to ↑ arthritis at adjacent joints and to pressure on nerves, leading to neurological compression syndromes and, when it occurs in the skull base, sensorineural deafness.”

“Paget’s disease is present in about 2% of the UK population over the age of 55. It’s prevalence increases with age, and it is more common in ♂ than ♀. Only about 10% of affected patients will have symptomatic disease. […] Most notable feature is pain. […] The diagnosis of Paget’s disease is primarily radiological. […] An isotope bone scan is frequently helpful in assessing the extent of skeletal involvement […] Deafness is present in up to half of cases of skull base Paget’s. • Other neurological complications are rare. […] Osteogenic sarcoma [is a] very rare complication of Paget’s disease. […] Any increase of pain in a patient with Paget’s disease should arouse suspicion of sarcomatous degeneration. A more common cause, however, is resumption of activity of disease. […] Treatment with agents that decrease bone turnover reduces disease activity […] Although such treatment has been shown to help pain, there is little evidence that it benefits other consequences of Paget’s disease. In particular, the deafness of Paget’s disease does not regress after treatment […] Bisphosphonates have become the mainstay of treatment. […] Goals of treatment [are to:] • Minimize symptoms. • Prevent long-term complications. • Normalize bone turnover. • Alkaline phosphatase in normal range. • No actual evidence that treatment achieves this.”

The rest of this post will be devoted to covering topics from chapter 8:

Neuroendocrine cells are found in many sites throughout the body. They are particularly prominent in the GI tract and pancreas and […] have the ability to synthesize, store, and release peptide hormones. […] the majority of neuroendocrine tumours occur within the gastroenteropancreatic axis. […] >50% are traditionally termed carcinoid tumours […] with the remainder largely comprising pancreatic islet cell tumours. • Carcinoid and islet cell tumours are generally slow-growing. […] There is a move towards standardizing the terminology of these tumours […] The term NEN [neuroendocrine neoplasia] included low- and intermediate-grade neoplasia (previously referred to as carcinoid or atypical carcinoid) which are now referred to as neuroendocrine tumours (NETs) and high-grade neoplasia (neuroendocrine carcinoma, NEC). There is a confusing array of classifications of NENs, based on anatomical origin, histology, and secretory activity. • Many of these classifications are well established and widely used.”

“It is important to understand the differences between ‘differentiation’, which is the extent to which the neoplastic cells resemble their non-tumourous counterparts, and ‘grade’, which is the inherent agressiveness of the tumour. […] Neuroendocrine carcinomas are the most aggressive NENs and can be either small or large cell type. […] NENs are diagnosed based on histological features of biopsy specimens. The presenting features of the tumours vary like any other tumour, based on their anatomical location, such as abdominal pain, intestinal obstruction. Many are incidentally discovered during endoscopy or imaging for unrelated conditions. In a database study, 49% of NENs were localized, 24% had regional metastases, and 27% had distant metastases. […] These tumours rarely manifest themselves due to their secretory effect. [This is quite different from some of the other tumours they covered elsewhere in the book – US]  [….] Only a third of patients with neuroendocrine tumours develop symptoms due to hormone secretion.”

“Surgery is the treatment of choice for NENs grades 1 and 2, except in the presence of widespread distant metastases and extensive local invasion. […] Somatostatin analogues (SSA) have relatively minor side effects and provide long-term symptom control. •Octreotide and lanreotide […] reduce the level of biochemical tumour markers in the majority of patients and control symptoms in around 70% of cases. […] A combination of interferon with octreotide has been shown to produce biochemical and symptomatic improvement in patients who have previously had no significant benefit from either drug alone. […] Cytotoxic chemotherapy may be considered in patients with progressive, advanced, or uncontrolled symptomatic disease.”

“Despite the changes in nomenclature of NENs […] the ‘carcinoid crisis’ [apparently also termed ‘malignant carcinoid syndrome‘, US] is still an important descriptive term. It is a potentially life-threatening condition that should be prevented, where possible, and treated as an emergency. • Clinical features include hypotension, tachycardia, arrhythmias, flushing, diarrhoea, broncospasm, and altered sensoriom. […] carcinoid crisis can be triggered by manipulation of the tumours, such as during biopsy, surgery, or palpation. • These result in the release of biologically active compounds from the tumours. […] Carcinoid heart disease […] result in valvular stenosis or regurgitation and eventually heart failure. This condition is seen in 40-50% of patients with carcinoid syndrome and 3-4% of patients with neuroendocrine tumours”.

“An insulinoma is a functioning neuroendocrine tumour of the pancreas that causes hypoglycemia through inappropriate secretion of insulin. • Unlike other neuroendocrine tumours of the pancreas, more than 90% of insulinomas are benign. […] annual incidence of insulinomas is of the order of 1-2 per million population. […] The treatment of choice in all, but poor, surgical candidates is operative removal. […] In experienced surgical hands, the mortality is less than 1%. […] Following the removal of solitary insulinoma [>80% of cases], life expectancy is restored to normal. Malignant insulinomas, with metastases usually to the liver, have a natural history of years, rather than months, and may be controlled with medical therapy or specific antitumour therapy […] • Average 5-year survival estimated to be approximately 35% for malignant insulinomas. […] Gastrinomas are the most common functional malignant pancreatic endocrine tumours. […] The incidence of gastrinomas is 0.5-2/million population/year. […] Gastrin […] is the principal gut hormone stimulating gastric acid secretion. • The Zollinger-Ellison (ZE) syndrome is characterized by gastric acid oversecretion and manifests itself as severe peptic ulcer disease (PUD), gastro-oesophageal reflux, and diarrhoea. […] 10-year survival [in patients with gastrinomas] without liver metastases is 95%. […] Where there are diffuse metastases, […] a 10-year survival of approximately 15% [is observed].”

One of the things I was thinking about before deciding whether or not to blog this chapter was whether the (fortunately!) rare conditions encountered in the chapter really ‘deserved’ to be covered. Unlike what is the case for, say, breast cancer or colon cancer, most people won’t know someone who’ll die from malignant insulinoma. However although these conditions are very rare, I also can’t stop myself from thinking they’re also quite interesting, and I don’t care much about whether I know someone with a disease I’ve read about. And if you think these conditions are rare, well, for glucagonomas “The annual incidence is estimated at 1 per 20 million population”. These very rare conditions really serve as a reminder of how great our bodies are at dealing with all kinds of problems we’ve never even thought about. We don’t think about them precisely because a problem so rarely arises – but just now and then, well…

Let’s talk a little bit more about those glucagonomas:

“Glucagonomas are neuroendocrine tumours that usually arise from the α cells of the pancreas and produce the glucagonoma syndrome through the secretion of glucagon and other peptides derived from the preproglucagon gene. • The large majority of glucagonomas are malignant, but they are also very indolent tumours, and the diagnosis may be overlooked for many years. • Up to 90% of patients will have lymph node or liver metastases at the time of presentation. • They are classically associated with the rash of necrolytic migratory erythema. […] The characteristic rash [….] occurs in >70% of cases […] glucose intolerance is a frequent association (>90%). • Sustained gluconeogenesis also causes amino acid deficiencies and results in protein catabolism which can be associated with unrelenting weight loss in >60% of patients. • Glucagon has a direct suppressive effect on the bone marrow, resulting in a normochromic normocytic anaemia in almost all patients. […] Surgery is the only curative option, but the potential for a complete cure may be as low as 5%.”

“In 1958, Verner and Morrison1 first described a syndrome consisting of refractory watery diarrhoea and hypokalaemia, associated with a neuroendocrine tumour of the pancreas. • The syndrome of watery diarrhea, hypokalaemia and acidosis (WDHA) is due to secretion of vasoactive intestinal polypeptide (VIP). • Tumours that secrete VIP are known as VIPomas. VIPomas account for <10% of islet cell tumours and mainly occur as solitary tumours. >60% are malignant […] The most prominent symptom in most patients is profuse watery diarrhoea […] Surgery to remove the tumour is the treatment of first choice […] and may be curative in around 40% of patients. […] Somatostatin analogues produce effective symptomatic relief from the diarrhoea in most patients. Long-term use does not result in tumour regression. […] Chemotherapy […] has resulted in response rates of >30%.”

So by now we know that somatostatin analogues can provide symptom relief in a variety of contexts when you’re dealing with these conditions. But wait, what happens if you get a functional tumour of the cells that produce somatostatins? Will this mean that you just feel great all the time, or that you at least don’t have any symptoms of disease? Well, not exactly…

Somatostatinomas are very rare neuroendocrine tumours, occurring both in the pancreas and in the duodenum. • >60% are large tumours located in the head or body of the pancreas. • The clinical syndrome may be diagnosed late in the course of disease when metastatic spread to local lymph nodes and the liver has already occurred. […] • Glucose intolerance or frank diabetes mellitus may have been observed for many years prior to the diagnosis and retrospectively often represents the first clinical sign. It is probably due to the inhibitory effect of somatostatin on insulin secretion. • A high incidence of gallstones has been described similar to that seen as a side effect with long-term somatostatin analogue therapy. • Diarrhoea, steatorrhoea, and weight loss appear to be consistent clinical features […this despite the fact that you use the hormone produced by these tumours to manage diarrhea in other endocrine tumours – it’s stuff like this which makes these rare disorders far from boring to read about! US] and may be associated with inhibition of the exocrine pancreas by somatostatin.”

May 1, 2018 Posted by | Books, Cancer/oncology, Cardiology, Diabetes, Epidemiology, Medicine, Neurology, Pharmacology | Leave a comment

100 cases in surgery (II)

Below I have added some links and quotes related to the last half of the book’s coverage.

Ischemic rest pain. (“Rest pain indicates inadequate tissue perfusion. *Urgent investigation and treatment is required to salvage the limb. […] The material of choice for bypass grafting is autogenous vein. […] The long-term patency of prosthetic grafts is inferior compared with autogenous vein.”)
Deep vein thrombosis.
Lymphedema. (“In lymphoedema, the vast majority of patients (>90 per cent) are treated conservatively. […] Debulking operations […] are only considered for a selected few patients where the function of the limb is impaired or those with recurrent attacks of severe cellulitis.”)
Varicose veins. Trendelenburg Test. (“Surgery on the superficial venous system should be avoided in patients with an incompetent deep venous system.”)
Testicular Torsion.
Benign Prostatic Hyperplasia.
Acute pyelonephritis. (“In patients with recurrent infection in the urinary system, significant pathology needs excluding such as malignancy, urinary tract stone disease and abnormal urinary tract anatomy.”)
Renal cell carcinomavon Hippel-Lindau syndrome. (“Approximately one-quarter to one-third of patients with renal cell carcinomas have metastases at presentation. […] The classic presenting triad of loin pain, a mass and haematuria only occurs in about 10 per cent of patients. More commonly, one of these features appears in isolation.”)
Haematuria. (“When taking the history, it is important to elicit the following: •Visible or non-visible: duration of haematuria • Age: cancers are more common with increasing age •Sex: females more likely to have urinary tract infections• Location: during micturition, was the haematuria always present (indicative of renal, ureteric or bladder pathology) or was it only present initially (suggestive of anterior urethral pathology) or present at the end of the stream (posterior urethra, bladder neck)? •Pain: more often associated with infection/inflammation/calculi, whereas malignancy tends to be painless •Associated lower urinary tract symptoms that will be helpful in determining aetiology •History of trauma Travel abroad […] •Previous urological surgery/history/recent instrumentation/prostatic biopsy •Medication, e.g. anticoagulants •Family history •Occupational history, e.g. rubber/dye occupational hazards are risk factors for developing transitional carcinoma of the bladder […] •Smoking status: increased risk, particularly of bladder cancer •General status, e.g. weight loss, reduced appetite […] Anticoagulation can often unmask other pathology in the urinary tract. […] Patients on oral anticoagulation who develop haematuria still require investigation.”)
Urinary retention. (“Acute and chronic retention are usually differentiated by the presence or absence of pain. Acute retention is painful, unlike chronic retention, when the bladder accommodates the increase in volume over time.”)
Colles’ fracture/Distal Radius Fractures. (“In all fractures the distal neurological and vascular status should be assessed.”)
Osteoarthritis. (“Radiological evidence of osteoarthritis is common, with 80 per cent of individuals over 80 years demonstrating some evidence of the condition. […] The commonest symptoms are pain, a reduction in mobility, and deformity of the affected joint.”)
Simmonds’ test.
Patella fracture.
Dislocated shoulder.
Femur fracture. (“Fractured neck of the femur is a relatively common injury following a fall in the elderly population. The rate of hip fracture doubles every decade from the age of 50 years. There is a female preponderance of three to one. […] it is important to take a comprehensive history, concentrating on the mechanism of injury. It is incorrect to assume that all falls are mechanical; it is not uncommon to find that the cause of the fall is actually due to a urinary or chest infection or even a silent myocardial infarction.”)
The Ottawa Ankle Rules.
Septic arthritis.
Carpal tunnel syndrome. Tinel’s test. Phalen’s Test. (“It is important, when examining a patient with suspected carpal tunnel syndrome, to carefully examine their neck, shoulder, and axilla. […] the source of the neurological compression may be proximal to the carpal tunnel”)
Acute Compartment Syndrome. (“Within the limbs there are a number of myofascial compartments. These consist of muscles contained within a relatively fixed-volume structure, bounded by fascial layers and bone. After trauma the pressure in the myofascial compartment increases. This pressure may exceed the venous capillary pressure, resulting in a loss of venous outflow from the compartment. The failure to clear metabolites also leads to the accumulation of fluid as a result of osmosis. If left untreated, the pressure will eventually exceed arterial pressure, leading to significant tissue ischaemia. The damage is irreversible after 4–6 h. Tibial fractures are the commonest cause of an acute compartment syndrome, which is thought to complicate up to 20 per cent of these injuries. […] The classical description of ‘pain out of proportion to the injury’ may [unfortunately] be difficult to determine if the clinician is inexperienced.”)
Hemarthrosis. (“Most knee injuries result in swelling which develops over hours rather than minutes. [A] history of immediate knee swelling suggests that there is a haemarthrosis.”)
Sickle cell crisis.
Cervical Spine Fracture. Neurogenic shock. NEXUS Criteria for C-Spine Imaging.
Slipped Capital Femoral Epiphysis. Trethowan sign. (“At any age, a limp in a child should always be taken seriously.”)

ATLS guidelines. (“The ATLS protocol should be followed even in the presence of obvious limb deformity, to ensure a potentially life-threatening injury is not missed.”)
Peritonsillar Abscess.
Epistaxis. Little’s area.
Croup. Acute epiglottitis. (“Acute epiglottitis is an absolute emergency and is usually caused by Haemophilus influenzae. There is significant swelling, and any attempt to examine the throat may result in airway obstruction. […] In adults it tends to cause a supraglottitis. It has a rapid progression and can lead to total airway obstruction. […] Stridor is an ominous sign and needs to be taken seriously.”)
Bell’s palsy.
Subarachnoid hemorrhageInternational subarachnoid aneurysm trial.
Chronic subdural hematoma. (“This condition is twice as common in men as women. Risk factors include chronic alcoholism, epilepsy, anticoagulant therapy (including aspirin) and thrombocytopenia. CSDH is more common in elderly patients due to cerebral atrophy. […] Initial misdiagnosis is, unfortunately, quite common. […] a chronic subdural haematoma should be suspected in confused patients with a history of a fall.”)
Extradural Haematoma. Cushing response. (“A direct blow to the temporo-parietal area is the commonest cause of an extradural haematoma. The bleed is normally arterial in origin. In 85 per cent of cases there is an associated skull fracture that causes damage to the middle meningeal artery. […] This situation represents a neurosurgical emergency. Without urgent decompression the patient will die. Unlike the chronic subdural, which can be treated with Burr hole drainage, the more dense acute arterial haematoma requires a craniotomy in order to evacuate it.”)
Cauda equina syndromeNeurosurgery for Cauda Equina Syndrome.
ASA classification. (“Patients having an operation within 3 months of a myocardial infarction carry a 30 per cent risk of reinfarction or cardiac death. This drops to 5 per cent after 6 months. […] Patients with COPD have difficulty clearing secretions from the lungs during the postoperative period. They also have a higher risk of basal atelectasis and are more prone to chest infections. These factors in combination with postoperative pain (especially in thoracic or abdominal major surgery) make them prone to respiratory complications. […] Patients with diabetes have an increased risk of postoperative complications because of the presence of microvascular and macrovascular disease: •Atherosclerosis: ischaemic heart disease/peripheral vascular disease/cerebrovascular disease •Nephropathy: renal insufficiency […] •Autonomic neuropathy: gastroparesis, decreased bladder tone •Peripheral neuropathy: lower-extremity ulceration, infection, gangrene •Poor wound healingIncreased risk of infection Tight glycaemic control (6–10 mmol/L) and the prevention of hypoglycaemia are critical in preventing perioperative and postoperative complications. The patient with diabetes should be placed first on the operating list to avoid prolonged fasting.
“)
MalnutritionHartmann’s procedure. (“Malnutrition leads to delayed wound healing, reduced ventilatory capacity, reduced immunity and an increased risk of infection. […] The two main methods of feeding are either by the enteral route or the parenteral route. Enteral feeding is via the gastrointestinal tract. It is less expensive and is associated with fewer complications than feeding by the parenteral route. […] The parenteral route should only be used if there is an inability to ingest, digest, absorb or propulse nutrients through the gastrointestinal tract. It can be administered by either a peripheral or central line. Peripheral parenteral nutrition can cause thrombophlebitis […] Sepsis is the most frequent and serious complication of centrally administered parenteral nutrition.”)
Acute Kidney Injury. (“The aetiology of acute renal failure can be thought of in three main categories: •Pre-renal: the glomerular filtration is reduced because of poor renal perfusion. This is usually caused by hypovolaemia as a result of acute blood loss, fluid depletion or hypotension. […] • Renal: this is the result of damage directly to the glomerulus or tubule. The use of drugs such as NSAIDs, contrast agents or aminoglycosides all have direct nephrotoxic effects. Acute tubular necrosis can occur as a result of prolonged hypoperfusion […]. Pre-existing renal disease such as diabetic nephropathy or glomerulonephritis makes patients more susceptible to further renal injury. •Post-renal: this can be simply the result of a blocked catheter. […] Calculi, blood clots, ureteric ligation and prostatic hypertrophy can also all lead to obstruction of urinary flow.”)
Post-operative ileus.

Pulmonary embolism.

April 18, 2018 Posted by | Books, Cancer/oncology, Cardiology, Gastroenterology, Infectious disease, Medicine, Nephrology, Neurology | Leave a comment

A few (more) diabetes papers of interest

Earlier this week I covered a couple of papers, but the second paper turned out to include a lot of interesting stuff so I decided to cut the post short and postpone my coverage of the other papers I’d intended to cover in that post until a later point in time; this post includes some of those other papers I’d intended to cover in that post.

i. TCF7L2 Genetic Variants Contribute to Phenotypic Heterogeneity of Type 1 Diabetes.

“Although the autoimmune destruction of β-cells has a major role in the development of type 1 diabetes, there is growing evidence that the differences in clinical, metabolic, immunologic, and genetic characteristics among patients (1) likely reflect diverse etiology and pathogenesis (2). Factors that govern this heterogeneity are poorly understood, yet these may have important implications for prognosis, therapy, and prevention.

The transcription factor 7 like 2 (TCF7L2) locus contains the single nucleotide polymorphism (SNP) most strongly associated with type 2 diabetes risk, with an ∼30% increase per risk allele (3). In a U.S. cohort, heterozygous and homozygous carriers of the at-risk alleles comprised 40.6% and 7.9%, respectively, of the control subjects and 44.3% and 18.3%, respectively, of the individuals with type 2 diabetes (3). The locus has no known association with type 1 diabetes overall (48), with conflicting reports in latent autoimmune diabetes in adults (816). […] Our studies in two separate cohorts have shown that the type 2 diabetes–associated TCF7L2 genetic variant is more frequent among specific subsets of individuals with autoimmune type 1 diabetes, specifically those with fewer markers of islet autoimmunity (22,23). These observations support a role of this genetic variant in the pathogenesis of diabetes at least in a subset of individuals with autoimmune diabetes. However, whether individuals with type 1 diabetes and this genetic variant have distinct metabolic abnormalities has not been investigated. We aimed to study the immunologic and metabolic characteristics of individuals with type 1 diabetes who carry a type 2 diabetes–associated allele of the TCF7L2 locus.”

“We studied 810 TrialNet participants with newly diagnosed type 1 diabetes and found that among individuals 12 years and older, the type 2 diabetes–associated TCF7L2 genetic variant is more frequent in those presenting with a single autoantibody than in participants who had multiple autoantibodies. These TCF7L2 variants were also associated with higher mean C-peptide AUC and lower mean glucose AUC levels at the onset of type 1 diabetes. […] These findings suggest that, besides the well-known link with type 2 diabetes, the TCF7L2 locus may play a role in the development of type 1 diabetes. The type 2 diabetes–associated TCF7L2 genetic variant identifies a subset of individuals with autoimmune type 1 diabetes and fewer markers of islet autoimmunity, lower glucose, and higher C-peptide at diagnosis. […] A possible interpretation of these data is that TCF7L2-encoded diabetogenic mechanisms may contribute to diabetes development in individuals with limited autoimmunity […]. Because the risk of progression to type 1 diabetes is lower in individuals with single compared with multiple autoantibodies, it is possible that in the absence of this type 2 diabetes–associated TCF7L2 variant, these individuals may have not manifested diabetes. If that is the case, we would postulate that disease development in these patients may have a type 2 diabetes–like pathogenesis in which islet autoimmunity is a significant component but not necessarily the primary driver.”

“The association between this genetic variant and single autoantibody positivity was present in individuals 12 years or older but not in children younger than 12 years. […] The results in the current study suggest that the type 2 diabetes–associated TCF7L2 genetic variant plays a larger role in older individuals. There is mounting evidence that the pathogenesis of type 1 diabetes varies by age (31). Younger individuals appear to have a more aggressive form of disease, with faster decline of β-cell function before and after onset of disease, higher frequency and severity of diabetic ketoacidosis, which is a clinical correlate of severe insulin deficiency, and lower C-peptide at presentation (3135). Furthermore, older patients are less likely to have type 1 diabetes–associated HLA alleles and islet autoantibodies (28). […] Taken together, we have demonstrated that individuals with autoimmune type 1 diabetes who carry the type 2 diabetes–associated TCF7L2 genetic variant have a distinct phenotype characterized by milder immunologic and metabolic characteristics than noncarriers, closer to those of type 2 diabetes, with an important effect of age.”

ii. Heart Failure: The Most Important, Preventable, and Treatable Cardiovascular Complication of Type 2 Diabetes.

“Concerns about cardiovascular disease in type 2 diabetes have traditionally focused on atherosclerotic vasculo-occlusive events, such as myocardial infarction, stroke, and limb ischemia. However, one of the earliest, most common, and most serious cardiovascular disorders in patients with diabetes is heart failure (1). Following its onset, patients experience a striking deterioration in their clinical course, which is marked by frequent hospitalizations and eventually death. Many sudden deaths in diabetes are related to underlying ventricular dysfunction rather than a new ischemic event. […] Heart failure and diabetes are linked pathophysiologically. Type 2 diabetes and heart failure are each characterized by insulin resistance and are accompanied by the activation of neurohormonal systems (norepinephrine, angiotensin II, aldosterone, and neprilysin) (3). The two disorders overlap; diabetes is present in 35–45% of patients with chronic heart failure, whether they have a reduced or preserved ejection fraction.”

“Treatments that lower blood glucose do not exert any consistently favorable effect on the risk of heart failure in patients with diabetes (6). In contrast, treatments that increase insulin signaling are accompanied by an increased risk of heart failure. Insulin use is independently associated with an enhanced likelihood of heart failure (7). Thiazolidinediones promote insulin signaling and have increased the risk of heart failure in controlled clinical trials (6). With respect to incretin-based secretagogues, liraglutide increases the clinical instability of patients with existing heart failure (8,9), and the dipeptidyl peptidase 4 inhibitors saxagliptin and alogliptin are associated with an increased risk of heart failure in diabetes (10). The likelihood of heart failure with the use of sulfonylureas may be comparable to that with thiazolidinediones (11). Interestingly, the only two classes of drugs that ameliorate hyperinsulinemia (metformin and sodium–glucose cotransporter 2 inhibitors) are also the only two classes of antidiabetes drugs that appear to reduce the risk of heart failure and its adverse consequences (12,13). These findings are consistent with experimental evidence that insulin exerts adverse effects on the heart and kidneys that can contribute to heart failure (14). Therefore, physicians can prevent many cases of heart failure in type 2 diabetes by careful consideration of the choice of agents used to achieve glycemic control. Importantly, these decisions have an immediate effect; changes in risk are seen within the first few months of changes in treatment. This immediacy stands in contrast to the years of therapy required to see a benefit of antidiabetes drugs on microvascular risk.”

“As reported by van den Berge et al. (4), the prognosis of patients with heart failure has improved over the past two decades; heart failure with a reduced ejection fraction is a treatable disease. Inhibitors of the renin-angiotensin system are a cornerstone of the management of both disorders; they prevent the onset of heart failure and the progression of nephropathy in patients with diabetes, and they reduce the risk of cardiovascular death and hospitalization in those with established heart failure (3,15). Diabetes does not influence the magnitude of the relative benefit of ACE inhibitors in patients with heart failure, but patients with diabetes experience a greater absolute benefit from treatment (16).”

“The totality of evidence from randomized trials […] demonstrates that in patients with diabetes, heart failure is not only common and clinically important, but it can also be prevented and treated. This conclusion is particularly significant because physicians have long ignored heart failure in their focus on glycemic control and their concerns about the ischemic macrovascular complications of diabetes (1).”

iii. Closely related to the above study: Mortality Reduction Associated With β-Adrenoceptor Inhibition in Chronic Heart Failure Is Greater in Patients With Diabetes.

“Diabetes increases mortality in patients with chronic heart failure (CHF) and reduced left ventricular ejection fraction. Studies have questioned the safety of β-adrenoceptor blockers (β-blockers) in some patients with diabetes and reduced left ventricular ejection fraction. We examined whether β-blockers and ACE inhibitors (ACEIs) are associated with differential effects on mortality in CHF patients with and without diabetes. […] We conducted a prospective cohort study of 1,797 patients with CHF recruited between 2006 and 2014, with mean follow-up of 4 years.”

RESULTS Patients with diabetes were prescribed larger doses of β-blockers and ACEIs than were patients without diabetes. Increasing β-blocker dose was associated with lower mortality in patients with diabetes (8.9% per mg/day; 95% CI 5–12.6) and without diabetes (3.5% per mg/day; 95% CI 0.7–6.3), although the effect was larger in people with diabetes (interaction P = 0.027). Increasing ACEI dose was associated with lower mortality in patients with diabetes (5.9% per mg/day; 95% CI 2.5–9.2) and without diabetes (5.1% per mg/day; 95% CI 2.6–7.6), with similar effect size in these groups (interaction P = 0.76).”

“Our most important findings are:

  • Higher-dose β-blockers are associated with lower mortality in patients with CHF and LVSD, but patients with diabetes may derive more benefit from higher-dose β-blockers.

  • Higher-dose ACEIs were associated with comparable mortality reduction in people with and without diabetes.

  • The association between higher β-blocker dose and reduced mortality is most pronounced in patients with diabetes who have more severely impaired left ventricular function.

  • Among patients with diabetes, the relationship between β-blocker dose and mortality was not associated with glycemic control or insulin therapy.”

“We make the important observation that patients with diabetes may derive more prognostic benefit from higher β-blocker doses than patients without diabetes. These data should provide reassurance to patients and health care providers and encourage careful but determined uptitration of β-blockers in this high-risk group of patients.”

iv. Diabetes, Prediabetes, and Brain Volumes and Subclinical Cerebrovascular Disease on MRI: The Atherosclerosis Risk in Communities Neurocognitive Study (ARIC-NCS).

“Diabetes and prediabetes are associated with accelerated cognitive decline (1), and diabetes is associated with an approximately twofold increased risk of dementia (2). Subclinical brain pathology, as defined by small vessel disease (lacunar infarcts, white matter hyperintensities [WMH], and microhemorrhages), large vessel disease (cortical infarcts), and smaller brain volumes also are associated with an increased risk of cognitive decline and dementia (37). The mechanisms by which diabetes contributes to accelerated cognitive decline and dementia are not fully understood, but contributions of hyperglycemia to both cerebrovascular disease and primary neurodegenerative disease have been suggested in the literature, although results are inconsistent (2,8). Given that diabetes is a vascular risk factor, brain atrophy among individuals with diabetes may be driven by increased cerebrovascular disease. Brain magnetic resonance imaging (MRI) provides a noninvasive opportunity to study associations of hyperglycemia with small vessel disease (lacunar infarcts, WMH, microhemorrhages), large vessel disease (cortical infarcts), and brain volumes (9).”

“Overall, the mean age of participants [(n = 1,713)] was 75 years, 60% were women, 27% were black, 30% had prediabetes (HbA1c 5.7 to <6.5%), and 35% had diabetes. Compared with participants without diabetes and HbA1c <5.7%, those with prediabetes (HbA1c 5.7 to <6.5%) were of similar age (75.2 vs. 75.0 years; P = 0.551), were more likely to be black (24% vs. 11%; P < 0.001), have less than a high school education (11% vs. 7%; P = 0.017), and have hypertension (71% vs. 63%; P = 0.012) (Table 1). Among participants with diabetes, those with HbA1c <7.0% versus ≥7.0% were of similar age (75.4 vs. 75.1 years; P = 0.481), but those with diabetes and HbA1c ≥7.0% were more likely to be black (39% vs. 28%; P = 0.020) and to have less than a high school education (23% vs. 16%; P = 0.031) and were more likely to have a longer duration of diabetes (12 vs. 8 years; P < 0.001).”

“Compared with participants without diabetes and HbA1c <5.7%, those with diabetes and HbA1c ≥7.0% had smaller total brain volume (β −0.20 SDs; 95% CI −0.31, −0.09) and smaller regional brain volumes, including frontal, temporal, occipital, and parietal lobes; deep gray matter; Alzheimer disease signature region; and hippocampus (all P < 0.05) […]. Compared with participants with diabetes and HbA1c <7.0%, those with diabetes and HbA1c ≥7.0% had smaller total brain volume (P < 0.001), frontal lobe volume (P = 0.012), temporal lobe volume (P = 0.012), occipital lobe volume (P = 0.008), parietal lobe volume (P = 0.015), deep gray matter volume (P < 0.001), Alzheimer disease signature region volume (0.031), and hippocampal volume (P = 0.016). Both participants with diabetes and HbA1c <7.0% and those with prediabetes (HbA1c 5.7 to <6.5%) had similar total and regional brain volumes compared with participants without diabetes and HbA1c <5.7% (all P > 0.05). […] No differences in the presence of lobar microhemorrhages, subcortical microhemorrhages, cortical infarcts, and lacunar infarcts were observed among the diabetes-HbA1c categories (all P > 0.05) […]. Compared with participants without diabetes and HbA1c <5.7%, those with diabetes and HbA1c ≥7.0% had increased WMH volume (P = 0.016). The WMH volume among participants with diabetes and HbA1c ≥7.0% was also significantly greater than among those with diabetes and HbA1c <7.0% (P = 0.017).”

“Those with diabetes duration ≥10 years were older than those with diabetes duration <10 years (75.9 vs. 75.0 years; P = 0.041) but were similar in terms of race and sex […]. Compared with participants with diabetes duration <10 years, those with diabetes duration ≥10 years has smaller adjusted total brain volume (β −0.13 SDs; 95% CI −0.20, −0.05) and smaller temporal lobe (β −0.14 SDs; 95% CI −0.24, −0.03), parietal lobe (β − 0.11 SDs; 95% CI −0.21, −0.01), and hippocampal (β −0.16 SDs; 95% CI −0.30, −0.02) volumes […]. Participants with diabetes duration ≥10 years also had a 2.44 times increased odds (95% CI 1.46, 4.05) of lacunar infarcts compared with those with diabetes duration <10 years”.

Conclusions
In this community-based population, we found that ARIC-NCS participants with diabetes with HbA1c ≥7.0% have smaller total and regional brain volumes and an increased burden of WMH, but those with prediabetes (HbA1c 5.7 to <6.5%) and diabetes with HbA1c <7.0% have brain volumes and markers of subclinical cerebrovascular disease similar to those without diabetes. Furthermore, among participants with diabetes, those with more-severe disease (as measured by higher HbA1c and longer disease duration) had smaller total and regional brain volumes and an increased burden of cerebrovascular disease compared with those with lower HbA1c and shorter disease duration. However, we found no evidence that associations of diabetes with smaller brain volumes are mediated by cerebrovascular disease.

The findings of this study extend the current literature that suggests that diabetes is strongly associated with brain volume loss (11,2527). Global brain volume loss (11,2527) has been consistently reported, but associations of diabetes with smaller specific brain regions have been less robust (27,28). Similar to prior studies, the current results show that compared with individuals without diabetes, those with diabetes have smaller total brain volume (11,2527) and regional brain volumes, including frontal and occipital lobes, deep gray matter, and the hippocampus (25,27). Furthermore, the current study suggests that greater severity of disease (as measured by HbA1c and diabetes duration) is associated with smaller total and regional brain volumes. […] Mechanisms whereby diabetes may contribute to brain volume loss include accelerated amyloid-β and hyperphosphorylated tau deposition as a result of hyperglycemia (29). Another possible mechanism involves pancreatic amyloid (amylin) infiltration of the brain, which then promotes amyloid-β deposition (29). […] Taken together, […] the current results suggest that diabetes is associated with both lower brain volumes and increased cerebrovascular pathology (WMH and lacunes).”

v. Interventions to increase attendance for diabetic retinopathy screening (Cochrane review).

“The primary objective of the review was to assess the effectiveness of quality improvement (QI) interventions that seek to increase attendance for DRS in people with type 1 and type 2 diabetes.

Secondary objectives were:
To use validated taxonomies of QI intervention strategies and behaviour change techniques (BCTs) to code the description of interventions in the included studies and determine whether interventions that include particular QI strategies or component BCTs are more effective in increasing screening attendance;
To explore heterogeneity in effect size within and between studies to identify potential explanatory factors for variability in effect size;
To explore differential effects in subgroups to provide information on how equity of screening attendance could be improved;
To critically appraise and summarise current evidence on the resource use, costs and cost effectiveness.”

“We included 66 RCTs conducted predominantly (62%) in the USA. Overall we judged the trials to be at low or unclear risk of bias. QI strategies were multifaceted and targeted patients, healthcare professionals or healthcare systems. Fifty-six studies (329,164 participants) compared intervention versus usual care (median duration of follow-up 12 months). Overall, DRS [diabetic retinopathy screening] attendance increased by 12% (risk difference (RD) 0.12, 95% confidence interval (CI) 0.10 to 0.14; low-certainty evidence) compared with usual care, with substantial heterogeneity in effect size. Both DRS-targeted (RD 0.17, 95% CI 0.11 to 0.22) and general QI interventions (RD 0.12, 95% CI 0.09 to 0.15) were effective, particularly where baseline DRS attendance was low. All BCT combinations were associated with significant improvements, particularly in those with poor attendance. We found higher effect estimates in subgroup analyses for the BCTs ‘goal setting (outcome)’ (RD 0.26, 95% CI 0.16 to 0.36) and ‘feedback on outcomes of behaviour’ (RD 0.22, 95% CI 0.15 to 0.29) in interventions targeting patients, and ‘restructuring the social environment’ (RD 0.19, 95% CI 0.12 to 0.26) and ‘credible source’ (RD 0.16, 95% CI 0.08 to 0.24) in interventions targeting healthcare professionals.”

“Ten studies (23,715 participants) compared a more intensive (stepped) intervention versus a less intensive intervention. In these studies DRS attendance increased by 5% (RD 0.05, 95% CI 0.02 to 0.09; moderate-certainty evidence).”

“Overall, we found that there is insufficient evidence to draw robust conclusions about the relative cost effectiveness of the interventions compared to each other or against usual care.”

“The results of this review provide evidence that QI interventions targeting patients, healthcare professionals or the healthcare system are associated with meaningful improvements in DRS attendance compared to usual care. There was no statistically significant difference between interventions specifically aimed at DRS and those which were part of a general QI strategy for improving diabetes care.”

vi. Diabetes in China: Epidemiology and Genetic Risk Factors and Their Clinical Utility in Personalized Medication.

“The incidence of type 2 diabetes (T2D) has rapidly increased over recent decades, and T2D has become a leading public health challenge in China. Compared with European descents, Chinese patients with T2D are diagnosed at a relatively young age and low BMI. A better understanding of the factors contributing to the diabetes epidemic is crucial for determining future prevention and intervention programs. In addition to environmental factors, genetic factors contribute substantially to the development of T2D. To date, more than 100 susceptibility loci for T2D have been identified. Individually, most T2D genetic variants have a small effect size (10–20% increased risk for T2D per risk allele); however, a genetic risk score that combines multiple T2D loci could be used to predict the risk of T2D and to identify individuals who are at a high risk. […] In this article, we review the epidemiological trends and recent progress in the understanding of T2D genetic etiology and further discuss personalized medicine involved in the treatment of T2D.”

“Over the past three decades, the prevalence of diabetes in China has sharply increased. The prevalence of diabetes was reported to be less than 1% in 1980 (2), 5.5% in 2001 (3), 9.7% in 2008 (4), and 10.9% in 2013, according to the latest published nationwide survey (5) […]. The prevalence of diabetes was higher in the senior population, men, urban residents, individuals living in economically developed areas, and overweight and obese individuals. The estimated prevalence of prediabetes in 2013 was 35.7%, which was much higher than the estimate of 15.5% in the 2008 survey. Similarly, the prevalence of prediabetes was higher in the senior population, men, and overweight and obese individuals. However, prediabetes was more prevalent in rural residents than in urban residents. […] the 2013 survey also compared the prevalence of diabetes among different races. The crude prevalence of diabetes was 14.7% in the majority group, i.e., Chinese Han, which was higher than that in most minority ethnic groups, including Tibetan, Zhuang, Uyghur, and Muslim. The crude prevalence of prediabetes was also higher in the Chinese Han ethnic group. The Tibetan participants had the lowest prevalence of diabetes and prediabetes (4.3% and 31.3%).”

“[T]he prevalence of diabetes in young people is relatively high and increasing. The prevalence of diabetes in the 20- to 39-year age-group was 3.2%, according to the 2008 national survey (4), and was 5.9%, according to the 2013 national survey (5). The prevalence of prediabetes also increased from 9.0% in 2008 to 28.8% in 2013 […]. Young people suffering from diabetes have a higher risk of chronic complications, which are the major cause of mortality and morbidity in diabetes. According to a study conducted in Asia (6), patients with young-onset diabetes had higher mean concentrations of HbA1c and LDL cholesterol and a higher prevalence of retinopathy (20% vs. 18%, P = 0.011) than those with late-onset diabetes. In the Chinese, patients with early-onset diabetes had a higher risk of nonfatal cardiovascular disease (7) than did patients with late-onset diabetes (odds ratio [OR] 1.91, 95% CI 1.81–2.02).”

“As approximately 95% of patients with diabetes in China have T2D, the rapid increase in the prevalence of diabetes in China may be attributed to the increasing rates of overweight and obesity and the reduction in physical activity, which is driven by economic development, lifestyle changes, and diet (3,11). According to a series of nationwide surveys conducted by the China Physical Fitness Surveillance Center (12), the prevalence of overweight (BMI ≥23.0 to <27.5 kg/m2) in Chinese adults aged 20–59 years increased from 37.4% in 2000 to 39.2% in 2005, 40.7% in 2010, and 41.2% in 2014, with an estimated increase of 0.27% per year. The prevalence of obesity (BMI ≥27.5 kg/m2) increased from 8.6% in 2000 to 10.3% in 2005, 12.2% in 2010, and 12.9% in 2014, with an estimated increase of 0.32% per year […]. The prevalence of central obesity increased from 13.9% in 2000 to 18.3% in 2005, 22.1% in 2010, and 24.9% in 2014, with an estimated increase of 0.78% per year. Notably, T2D develops at a considerably lower BMI in the Chinese population than that in European populations. […] The relatively high risk of diabetes at a lower BMI could be partially attributed to the tendency toward visceral adiposity in East Asian populations, including the Chinese population (13). Moreover, East Asian populations have been found to have a higher insulin sensitivity with a much lower insulin response than European descent and African populations, implying a lower compensatory β-cell function, which increases the risk of progressing to overt diabetes (14).”

“Over the past two decades, linkage analyses, candidate gene approaches, and large-scale GWAS have successfully identified more than 100 genes that confer susceptibility to T2D among the world’s major ethnic populations […], most of which were discovered in European populations. However, less than 50% of these European-derived loci have been successfully confirmed in East Asian populations. […] there is a need to identify specific genes that are associated with T2D in other ethnic populations. […] Although many genetic loci have been shown to confer susceptibility to T2D, the mechanism by which these loci participate in the pathogenesis of T2D remains unknown. Most T2D loci are located near genes that are related to β-cell function […] most single nucleotide polymorphisms (SNPs) contributing to the T2D risk are located in introns, but whether these SNPs directly modify gene expression or are involved in linkage disequilibrium with unknown causal variants remains to be investigated. Furthermore, the loci discovered thus far collectively account for less than 15% of the overall estimated genetic heritability.”

“The areas under the receiver operating characteristic curves (AUCs) are usually used to assess the discriminative accuracy of an approach. The AUC values range from 0.5 to 1.0, where an AUC of 0.5 represents a lack of discrimination and an AUC of 1 represents perfect discrimination. An AUC ≥0.75 is considered clinically useful. The dominant conventional risk factors, including age, sex, BMI, waist circumference, blood pressure, family history of diabetes, physical activity level, smoking status, and alcohol consumption, can be combined to construct conventional risk factor–based models (CRM). Several studies have compared the predictive capacities of models with and without genetic information. The addition of genetic markers to a CRM could slightly improve the predictive performance. For example, one European study showed that the addition of an 11-SNP GRS to a CRM marginally improved the risk prediction (AUC was 0.74 without and 0.75 with the genetic markers, P < 0.001) in a prospective cohort of 16,000 individuals (37). A meta-analysis (38) consisting of 23 studies investigating the predictive performance of T2D risk models also reported that the AUCs only slightly increased with the addition of genetic information to the CRM (median AUC was increased from 0.78 to 0.79). […] Despite great advances in genetic studies, the clinical utility of genetic information in the prediction, early identification, and prevention of T2D remains in its preliminary stage.”

“An increasing number of studies have highlighted that early nutrition has a persistent effect on the risk of diabetes in later life (40,41). China’s Great Famine of 1959–1962 is considered to be the largest and most severe famine of the 20th century […] Li et al. (43) found that offspring of mothers exposed to the Chinese famine have a 3.9-fold increased risk of diabetes or hyperglycemia as adults. A more recent study (the Survey on Prevalence in East China for Metabolic Diseases and Risk Factors [SPECT-China]) conducted in 2014, among 6,897 adults from Shanghai, Jiangxi, and Zhejiang provinces, had the same conclusion that famine exposure during the fetal period (OR 1.53, 95% CI 1.09–2.14) and childhood (OR 1.82, 95% CI 1.21–2.73) was associated with diabetes (44). These findings indicate that undernutrition during early life increases the risk of hyperglycemia in adulthood and this association is markedly exaggerated when facing overnutrition in later life.”

February 23, 2018 Posted by | Cardiology, Diabetes, Epidemiology, Genetics, Health Economics, Immunology, Medicine, Neurology, Ophthalmology, Pharmacology, Studies | Leave a comment

Endocrinology (part 5 – calcium and bone metabolism)

Some observations from chapter 6:

“*Osteoclasts – derived from the monocytic cells; resorb bone. *Osteoblasts – derived from the fibroblast-like cells; make bone. *Osteocytes – buried osteoblasts; sense mechanical strain in bone. […] In order to ensure that bone can undertake its mechanical and metabolic functions, it is in a constant state of turnover […] Bone is laid down rapidly during skeletal growth at puberty. Following this, there is a period of stabilization of bone mass in early adult life. After the age of ~40, there is a gradual loss of bone in both sexes. This occurs at the rate of approximately 0.5% annually. However, in ♀ after the menopause, there is a period of rapid bone loss. The accelerated loss is maximal in the first 2-5 years after the cessation of ovarian function and then gradually declines until the previous gradual rate of loss is once again established. The excess bone loss associated with the menopause is of the order of 10% of skeletal mass. This menopause-associated loss, coupled with higher peak bone mass acquisition in ♂, largely explains why osteoporosis and its associated fractures are more common in ♀.”

“The clinical utility of routine measurements of bone turnover markers is not yet established. […] Skeletal radiology[:] *Useful for: *Diagnosis of fracture. *Diagnosis of specific diseases (e.g. Paget’s disease and osteomalacia). *Identification of bone dysplasia. *Not useful for assessing bone density. […] Isotope bone scans are useful for identifying localized areas of bone disease, such as fracture, metastases, or Paget’s disease. […] Isotope bone scans are particularly useful in Paget’s disease to establish the extent and sites of skeletal involvement and the underlying disease activity. […] Bone biopsy is occasionally necessary for the diagnosis of patients with complex metabolic bone diseases. […] Bone biopsy is not indicated for the routine diagnosis of osteoporosis. It should only be undertaken in highly specialist centres with appropriate expertise. […] Measurement of 24h urinary excretion of calcium provides a measure of risk of renal stone formation or nephrocalcinosis in states of chronic hypercalcaemia. […] 250H vitamin D […] is the main storage form of vitamin D, and the measurement of ‘total vitamin D’ is the most clinically useful measure of vitamin D status. Internationally, there remains controversy around a ‘normal’ or ‘optimal’ concentration of vitamin D. Levels over 50nmol/L are generally accepted as satisfactory and values <25nmol/L representing deficiency. True osteomalacia occurs with vitamin D values <15 nmol/L. Low levels of 250HD can result from a variety of causes […] Bone mass is quoted in terms of the number of standard deviations from an expected mean. […] A reduction of one SD in bone density will approximately double the risk of fracture.”

[I should perhaps add a cautionary note here that while this variable is very useful in general, it is more useful in some contexts than in others; and in some specific disease process contexts it is quite clear that it will tend to underestimate the fracture risk. Type 1 diabetes is a clear example. For more details, see this post.]

“Hypercalcaemia is found in 5% of hospital patients and in 0.5% of the general population. […] Many different disease states can lead to hypercalcaemia. […] In asymptomatic community-dwelling subjects, the vast majority of hypercalcaemia is the result of hyperparathyroidism. […] The clinical features of hypercalcaemia are well recognized […]; unfortunately, they are non-specific […] [They include:] *Polyuria. *Polydipsia. […] *Anorexia. *Vomiting. *Constipation. *Abdominal pain. […] *Confusion. *Lethargy. *Depression. […] Clinical signs of hypercalcaemia are rare. […] the presence of bone pain or fracture and renal stones […] indicate the presence of chronic hypercalcaemia. […] Hypercalcaemia is usually a late manifestation of malignant disease, and the primary lesion is usually evident by the time hypercalcaemia is expressed (50% of patients die within 30 days).”

“Primary hyperparathyroidism [is] [p]resent in up to 1 in 500 of the general population where it is predominantly a disease of post-menopausal ♀ […] The normal physiological response to hypocalcaemia is an increase in PTH secretion. This is termed 2° hyperparathyroidism and is not pathological in as much as the PTH secretion remains under feedback control. Continued stimulation of the parathyroid glands can lead to autonomous production of PTH. This, in turn, causes hypercalcaemia which is termed tertiary hyperparathyroidism. This is usually seen in the context of renal disease […] In majority of patients [with hyperparathyroidism] without end-organ damage, disease is benign and stable. […] Investigation is, therefore, primarily aimed at determining the presence of end-organ damage from hypercalcaemia in order to determine whether operative intervention is indicated. […] It is generally accepted that all patients with symptomatic hyperparathyroidism or evidence of end-organ damage should be considered for parathyroidectomy. This would include: *Definite symptoms of hypercalcaemia. […] *Impaired renal function. *Renal stones […] *Parathyroid bone disease, especially osteitis fibrosis cystica. *Pancreatitis. […] Patients not managed with surgery require regular follow-up. […] <5% fail to become normocalcaemic [after surgery], and these should be considered for a second operation. […] Patients rendered permanently hypoparathyroid by surgery require lifelong supplements of active metabolites of vitamin D with calcium. This can lead to hypercalciuria, and the risk of stone formation may still be present in these patients. […] In hypoparathyroidism, the target serum calcium should be at the low end of the reference range. […] any attempt to raise the plasma calcium well into the normal range is likely to result in unacceptable hypercalciuria”.

“Although hypocalcaemia can result from failure of any of the mechanisms by which serum calcium concentration is maintained, it is usually the result of either failure of PTH secretion or because of the inability to release calcium from bone. […] The clinical features of hypocalcaemia are largely as a result of neuromuscular excitability. In order of  severity, these include: *Tingling – especially of fingers, toes, or lips. *Numbness – especially of fingers, toes, or lips. *Cramps. *Carpopedal spasm. *Stridor due to laryngospasm. *Seizures. […] symptoms of hypocalcaemia tend to reflect the severity and rapidity of onset of the metabolic abnormality. […] there may be clinical signs and symptoms associated with the underlying condition: *Vitamin D deficiency may be associated with generalized bone pain, fractures, or proximal myopathy […] *Hypoparathyroidism can be accompanied by mental slowing and personality disturbances […] *If hypocalcaemia is present during the development of permanent teeth, these may show areas of enamel hypoplasia. This can be a useful physical sign, indicating that the hypocalcaemia is long-standing. […] Acute symptomatic hypocalcaemia is a medical emergency and demands urgent treatment whatever the cause […] *Patients with tetany or seizures require urgent IV treatment with calcium gluconate […] Care must be taken […] as too rapid elevation of the plasma calcium can cause arrhythmias. […] *Treatment of chronic hypocalcaemia is more dependent on the cause. […] In patients with mild parathyroid dysfunction, it may be possible to achieve acceptable calcium concentrations by using calcium supplements alone. […] The majority of patients will not achieve adequate control with such treatment. In those cases, it is necessary to use vitamin D or its metabolites in pharmacological doses to maintain plasma calcium.”

“Pseudohypoparathyroidism[:] *Resistance to parathyroid hormone action. *Due to defective signalling of PTH action via cell membrane receptor. *Also affects TSH, LH, FSH, and GH signalling. […] Patients with the most common type of pseudohypoparathyroidism (type 1a) have a characteristic set of skeletal abnormalities, known as Albright’s hereditary osteodystrophy. This comprises: *Short stature. *Obesity. *Round face. *Short metacarpals. […] The principles underlying the treatment of pseudohypoparathyroidism are the same as those underlying hypoparathyroidism. *Patients with the most common form of pseudohypoparathyroidism may have resistance to the action of other hormones which rely on G protein signalling. They, therefore, need to be assessed for thyroid and gonadal dysfunction (because of defective TSH and gonadotrophin action). If these deficiencies are present, they need to be treated in the conventional manner.”

“Osteomalacia occurs when there is inadequate mineralization of mature bone. Rickets is a disorder of the growing skeleton where there is inadequate mineralization of bone as it is laid down at the epiphysis. In most instances, osteomalacia leads to build-up of excessive unmineralized osteoid within the skeleton. In rickets, there is build-up of unmineralized osteoid in the growth plate. […] These two related conditions may coexist. […] Clinical features [of osteomalacia:] *Bone pain. *Deformity. *Fracture. *Proximal myopathy. *Hypocalcaemia (in vitamin D deficiency). […] The majority of patients with osteomalacia will show no specific radiological abnormalities. *The most characteristic abnormality is the Looser’s zone or pseudofracture. If these are present, they are virtually pathognomonic of osteomalacia. […] Oncogenic osteomalacia[:] Certain tumours appear to be able to produce FGF23 which is phosphaturic. This is rare […] Clinically, such patients usually present with profound myopathy as well as bone pain and fracture. […] Complete removal of the tumour results in resolution of the biochemical and skeletal abnormalities. If this is not possible […], treatment with vitamin D metabolites and phosphate supplements […] may help the skeletal symptoms.”

Hypophosphataemia[:] Phosphate is important for normal mineralization of bone. In the absence of sufficient phosphate, osteomalacia results. […] In addition, phosphate is important in its own right for neuromuscular function, and profound hypophosphataemia can be accompanied by encephalopathy, muscle weakness, and cardiomyopathy. It must be remembered that, as phosphate is primarily an intracellular anion, a low plasma phosphate does not necessarily represent actual phosphate depletion. […] Mainstay [of treatment] is phosphate replacement […] *Long-term administration of phosphate supplements stimulates parathyroid activity. This can lead to hypercalcaemia, a further fall in phosphate, with worsening of the bone disease […] To minimize parathyroid stimulation, it is usual to give one of the active metabolites of vitamin D in conjunction with phosphate.”

“Although the term osteoporosis refers to the reduction in the amount of bony tissue within the skeleton, this is generally associated with a loss of structural integrity of the internal architecture of the bone. The combination of both these changes means that osteoporotic bone is at high risk of fracture, even after trivial injury. […] Historically, there has been a primary reliance on bone mineral density as a threshold for treatment, whereas currently there is far greater emphasis on assessing individual patients’ risk of fracture that incorporates multiple clinical risk factors as well as bone mineral density. […] Osteoporosis may arise from a failure of the body to lay down sufficient bone during growth and maturation; an earlier than usual onset of bone loss following maturity; or an rate of that loss. […] Early menopause or late puberty (in ♂ or ♀) is associated with risk of osteoporosis. […] Lifestyle factors affecting bone mass [include:] *weight-bearing exercise [increase bone mass] […] *Smoking. *Excessive alcohol. *Nulliparity. *Poor calcium nutrition. [These all decrease bone mass] […] The risk of osteoporotic fracture increases with age. Fracture rates in ♂ are approximately half of those seen in ♀ of the same age. An ♀ aged 50 has approximately a 1:2 chance [risk, surely… – US] of sustaining an osteoporotic fracture in the rest of her life. The corresponding figure for a ♂ is 1:5. […] One-fifth of hip fracture victims will die within 6 months of the injury, and only 50% will return to their previous level of independence.”

“Any fracture, other than those affecting fingers, toes, or face, which is caused by a fall from standing height or less is called a fragility (low-trauma) fracture, and underlying osteoporosis should be considered. Patients suffering such a fracture should be considered for investigation and/or treatment for osteoporosis. […] [Osteoporosis is] [u]sually clinically silent until an acute fracture. *Two-thirds of vertebral fractures do not come to clinical attention. […] Osteoporotic vertebral fractures only rarely lead to neurological impairment. Any evidence of spinal cord compression should prompt a search for malignancy or other underlying cause. […] Osteoporosis does not cause generalized skeletal pain. […] Biochemical markers of bone turnover may be helpful in the calculation of fracture risk and in judging the response to drug therapies, but they have no role in the diagnosis of osteoporosis. […] An underlying cause for osteoporosis is present in approximately 10-30% of women and up to 50% of men with osteoporosis. […] 2° causes of osteoporosis are more common in ♂ and need to be excluded in all ♂ with osteoporotic fracture. […] Glucocorticoid treatment is one of the major 2° causes of osteoporosis.”

February 22, 2018 Posted by | Books, Cancer/oncology, Diabetes, Epidemiology, Medicine, Nephrology, Neurology, Pharmacology | Leave a comment

A few diabetes papers of interest

(I hadn’t expected to only cover two papers in this post, but the second paper turned out to include a lot of stuff I figured might be worth adding here. I might add another post later this week including some of the other studies I had intended to cover in this post.)

i. Burden of Mortality Attributable to Diagnosed Diabetes: A Nationwide Analysis Based on Claims Data From 65 Million People in Germany.

“Diabetes is among the 10 most common causes of death worldwide (2). Between 1990 and 2010, the number of deaths attributable to diabetes has doubled (2). People with diabetes have a reduced life expectancy of ∼5 to 6 years (3). The most common cause of death in people with diabetes is cardiovascular disease (3,4). Over the past few decades, a reduction of diabetes mortality has been observed in several countries (59). However, the excess risk of death is still higher than in the population without diabetes, particularly in younger age-groups (4,9,10). Unfortunately, in most countries worldwide, reliable data on diabetes mortality are lacking (1). In a few European countries, such as Denmark (5) and Sweden (4), mortality analyses are based on national diabetes registries that include all age-groups. However, Germany and many other European countries do not have such national registries. Until now, age-standardized hazard ratios for diabetes mortality between 1.4 and 2.6 have been published for Germany on the basis of regional studies and surveys with small respondent numbers (1114). To the best of our knowledge, no nationwide estimates of the number of excess deaths due to diabetes have been published for Germany, and no information on older age-groups >79 years is currently available.

In 2012, changes in the regulation of data transparency enabled the use of nationwide routine health care data from the German statutory health insurance system, which insures ∼90% of the German population (15). These changes have allowed for new possibilities for estimating the burden of diabetes in Germany. Hence, this study estimates the number of excess deaths due to diabetes (ICD-10 codes E10–E14) and type 2 diabetes (ICD-10 code E11) in Germany, which is the number of deaths that could have been prevented if the diabetes mortality rate was as high as that of the population without diabetes.”

“Nationwide data on mortality ratios for diabetes and no diabetes are not available for Germany. […] the age- and sex-specific mortality rate ratios between people with diabetes and without diabetes were used from a Danish study wherein the Danish National Diabetes Register was linked to the individual mortality data from the Civil Registration System that includes all people residing in Denmark (5). Because the Danish National Diabetes Register is one of the most accurate diabetes registries in Europe, with a sensitivity of 86% and positive predictive value of 90% (5), we are convinced that the Danish estimates are highly valid and reliable. Denmark and Germany have a comparable standard of living and health care system. The diabetes prevalence in these countries is similar (Denmark 7.2%, Germany 7.4% [20]) and mortality of people with and without diabetes comparable, as shown in the European mortality database”

“In total, 174,627 excess deaths (137,950 from type 2 diabetes) could have been prevented in 2010 if mortality was the same in people with and without diabetes. Overall, 21% of all deaths in Germany were attributable to diabetes, and 16% were attributable to type 2 diabetes […] Most of the excess deaths occurred in the 70- to 79- and 80- to 89-year-old age-groups (∼34% each) […]. Substantial sex differences were found in diabetes-related excess deaths. From the age of ∼40 years, the number of male excess deaths due to diabetes started to grow, but the number of female excess deaths increased with a delay. Thus, the highest number of male excess deaths due to diabetes occurred at the age of ∼75 years, whereas the peak of female excess deaths was ∼10 years later. […] The diabetes mortality rates increased with age and were always higher than in the population without diabetes. The largest differences in mortality rates between people with and without diabetes were observed in the younger age-groups. […] These results are in accordance with previous studies worldwide (3,4,7,9) and regional studies in Germany (1113).”

“According to official numbers from the Federal Statistical Office, 858,768 people died in Germany in 2010, with 23,131 deaths due to diabetes, representing 2.7% of the all-cause mortality (26). Hence, in Germany, diabetes is not ranked among the top 10 most common causes of death […]. We found that 21% of all deaths were attributable to diabetes and 16% were attributable to type 2 diabetes; hence, we suggest that the number of excess deaths attributable to diabetes is strongly underestimated if we rely on reported causes of death from death certificates, as official statistics do. Estimating diabetes-related mortality is challenging because most people die as a result of diabetes complications and comorbidities, such as cardiovascular disease and renal failure, which often are reported as the underlying cause of death (1,23). For this reason, another approach is to focus not only on the underlying cause of death but also on the multiple causes of death to assess any mention of a disease on the death certificate (27). In a study from Italy, the method of assessing multiple causes of death revealed that in 12.3% of all studied death certificates, diabetes was mentioned, whereas only 2.9% reported diabetes as the underlying cause of death (27), corresponding to a four times higher proportion of death related to diabetes. Another nationwide analysis from Canada found that diabetes was more than twice as likely to be a contributing factor to death than the underlying cause of death from the years 2004–2008 (28). A recently published study from the U.S. that was based on two representative surveys from 1997 to 2010 found that 11.5% of all deaths were attributable to diabetes, which reflects a three to four times higher proportion of diabetes-related deaths (29). Overall, these results, together with the current calculations, demonstrate that deaths due to diabetes contribute to a much higher burden than previously assumed.”

ii. Standardizing Clinically Meaningful Outcome Measures Beyond HbA1c for Type 1 Diabetes: A Consensus Report of the American Association of Clinical Endocrinologists, the American Association of Diabetes Educators, the American Diabetes Association, the Endocrine Society, JDRF International, The Leona M. and Harry B. Helmsley Charitable Trust, the Pediatric Endocrine Society, and the T1D Exchange.

“Type 1 diabetes is a life-threatening, autoimmune disease that strikes children and adults and can be fatal. People with type 1 diabetes have to test their blood glucose multiple times each day and dose insulin via injections or an infusion pump 24 h a day every day. Too much insulin can result in hypoglycemia, seizures, coma, or death. Hyperglycemia over time leads to kidney, heart, nerve, and eye damage. Even with diligent monitoring, the majority of people with type 1 diabetes do not achieve recommended target glucose levels. In the U.S., approximately one in five children and one in three adults meet hemoglobin A1c (HbA1c) targets and the average patient spends 7 h a day hyperglycemic and over 90 min hypoglycemic (13). […] HbA1c is a well-accepted surrogate outcome measure for evaluating the efficacy of diabetes therapies and technologies in clinical practice as well as in research (46). […] While HbA1c is used as a primary outcome to assess glycemic control and as a surrogate for risk of developing complications, it has limitations. As a measure of mean blood glucose over 2 or 3 months, HbA1c does not capture short-term variations in blood glucose or exposure to hypoglycemia and hyperglycemia in individuals with type 1 diabetes; HbA1c also does not capture the impact of blood glucose variations on individuals’ quality of life. Recent advances in type 1 diabetes technologies have made it feasible to assess the efficacy of therapies and technologies using a set of outcomes beyond HbA1c and to expand definitions of outcomes such as hypoglycemia. While definitions for hypoglycemia in clinical care exist, they have not been standardized […]. The lack of standard definitions impedes and can confuse their use in clinical practice, impedes development processes for new therapies, makes comparison of studies in the literature challenging, and may lead to regulatory and reimbursement decisions that fail to meet the needs of people with diabetes. To address this vital issue, the type 1 diabetes–stakeholder community launched the Type 1 Diabetes Outcomes Program to develop consensus definitions for a set of priority outcomes for type 1 diabetes. […] The outcomes prioritized under the program include hypoglycemia, hyperglycemia, time in range, diabetic ketoacidosis (DKA), and patient-reported outcomes (PROs).”

“Hypoglycemia is a significant — and potentially fatal — complication of type 1 diabetes management and has been found to be a barrier to achieving glycemic goals (9). Repeated exposure to severe hypoglycemic events has been associated with an increased risk of cardiovascular events and all-cause mortality in people with type 1 or type 2 diabetes (10,11). Hypoglycemia can also be fatal, and severe hypoglycemic events have been associated with increased mortality (1214). In addition to the physical aspects of hypoglycemia, it can also have negative consequences on emotional status and quality of life.

While there is some variability in how and when individuals manifest symptoms of hypoglycemia, beginning at blood glucose levels <70 mg/dL (3.9 mmol/L) (which is at the low end of the typical post-absorptive plasma glucose range), the body begins to increase its secretion of counterregulatory hormones including glucagon, epinephrine, cortisol, and growth hormone. The release of these hormones can cause moderate autonomic effects, including but not limited to shaking, palpitations, sweating, and hunger (15). Individuals without diabetes do not typically experience dangerously low blood glucose levels because of counterregulatory hormonal regulation of glycemia (16). However, in individuals with type 1 diabetes, there is often a deficiency of the counterregulatory response […]. Moreover, as people with diabetes experience an increased number of episodes of hypoglycemia, the risk of hypoglycemia unawareness, impaired glucose counterregulation (for example, in hypoglycemia-associated autonomic failure [17]), and level 2 and level 3 hypoglycemia […] all increase (18). Therefore, it is important to recognize and treat all hypoglycemic events in people with type 1 diabetes, particularly in populations (children, the elderly) that may not have the ability to recognize and self-treat hypoglycemia. […] More notable clinical symptoms begin at blood glucose levels <54 mg/dL (3.0 mmol/L) (19,20). As the body’s primary utilizer of glucose, the brain is particularly sensitive to decreases in blood glucose concentrations. Both experimental and clinical evidence has shown that, at these levels, neurogenic and neuroglycopenic symptoms including impairments in reaction times, information processing, psychomotor function, and executive function begin to emerge. These neurological symptoms correlate to altered brain activity in multiple brain areas including the prefrontal cortex and medial temporal lobe (2124). At these levels, individuals may experience confusion, dizziness, blurred or double vision, tremors, and tingling sensations (25). Hypoglycemia at this glycemic level may also increase proinflammatory and prothrombotic markers (26). Left untreated, these symptoms can become severe to the point that an individual will require assistance from others to move or function. Prolonged untreated hypoglycemia that continues to drop below 50 mg/dL (2.8 mmol/L) increases the risk of seizures, coma, and death (27,28). Hypoglycemia that affects cognition and stamina may also increase the risk of accidents and falls, which is a particular concern for older adults with diabetes (29,30).

The glycemic thresholds at which these symptoms occur, as well as the severity with which they manifest themselves, may vary in individuals with type 1 diabetes depending on the number of hypoglycemic episodes they have experienced (3133). Counterregulatory physiological responses may evolve in patients with type 1 diabetes who endure repeated hypoglycemia over time (34,35).”

“The Steering Committee defined three levels of hypoglycemia […] Level 1 hypoglycemia is defined as a measurable glucose concentration <70 mg/dL (3.9 mmol/L) but ≥54 mg/dL (3.0 mmol/L) that can alert a person to take action. A blood glucose concentration of 70 mg/dL (3.9 mmol/L) has been recognized as a marker of physiological hypoglycemia in humans, as it approximates the glycemic threshold for neuroendocrine responses to falling glucose levels in individuals without diabetes. As such, blood glucose in individuals without diabetes is generally 70–100 mg/dL (3.9–5.6 mmol/L) upon waking and 70–140 mg/dL (3.9–7.8 mmol/L) after meals, and any excursions beyond those levels are typically countered with physiological controls (16,37). However, individuals with diabetes who have impaired or altered counterregulatory hormonal and neurological responses do not have the same internal regulation as individuals without diabetes to avoid dropping below 70 mg/dL (3.9 mmol/L) and becoming hypoglycemic. Recurrent episodes of hypoglycemia lead to increased hypoglycemia unawareness, which can become dangerous as individuals cease to experience symptoms of hypoglycemia, allowing their blood glucose levels to continue falling. Therefore, glucose levels <70 mg/dL (3.9 mmol/L) are clinically important, independent of the severity of acute symptoms.

Level 2 hypoglycemia is defined as a measurable glucose concentration <54 mg/dL (3.0 mmol/L) that needs immediate action. At ∼54 mg/dL (3.0 mmol/L), neurogenic and neuroglycopenic hypoglycemic symptoms begin to occur, ultimately leading to brain dysfunction at levels <50 mg/dL (2.8 mmol/L) (19,20). […] Level 3 hypoglycemia is defined as a severe event characterized by altered mental and/or physical status requiring assistance. Severe hypoglycemia captures events during which the symptoms associated with hypoglycemia impact a patient to such a degree that the patient requires assistance from others (27,28). […] Hypoglycemia that sets in relatively rapidly, such as in the case of a significant insulin overdose, may induce level 2 or level 3 hypoglycemia with little warning (38).”

“The data regarding the effects of chronic hyperglycemia on long-term outcomes is conclusive, indicating that chronic hyperglycemia is a major contributor to morbidity and mortality in type 1 diabetes (41,4345). […] Although the correlation between long-term poor glucose control and type 1 diabetes complications is well established, the impact of short-term hyperglycemia is not as well understood. However, hyperglycemia has been shown to have physiological effects and in an acute-care setting is linked to morbidity and mortality in people with and without diabetes. Short-term hyperglycemia, regardless of diabetes diagnosis, has been shown to reduce survival rates among patients admitted to the hospital with stroke or myocardial infarction (47,48). In addition to increasing mortality, short-term hyperglycemia is correlated with stroke severity and poststroke disability (49,50).

The effects of short-term hyperglycemia have also been observed in nonacute settings. Evidence indicates that hyperglycemia alters retinal cell firing through sensitization in patients with type 1 diabetes (51). This finding is consistent with similar findings showing increased oxygen consumption and blood flow in the retina during hyperglycemia. Because retinal cells absorb glucose through an insulin-independent process, they respond more strongly to increases in glucose in the blood than other cells in patients with type 1 diabetes. The effects of acute hyperglycemia on retinal response may underlie part of the development of retinopathy known to be a long-term complication of type 1 diabetes.”

“The Steering Committee defines hyperglycemia for individuals with type 1 diabetes as the following:

  • Level 1—elevated glucose: glucose >180 mg/dL (10 mmol/L) and glucose ≤250 mg/dL (13.9 mmol/L)

  • Level 2—very elevated glucose: glucose >250 mg/dL (13.9 mmol/L) […]

Elevated glucose is defined as a glucose concentration >180 mg/dL (10.0 mmol/L) but ≤250 mg/dL (13.9 mmol/L). In clinical practice, measures of hyperglycemia differ based on time of day (e.g., pre- vs. postmeal). This program, however, focused on defining outcomes for use in product development that are universally applicable. Glucose profiles and postprandial blood glucose data for individuals without diabetes suggest that 140 mg/dL (7.8 mmol/L) is the appropriate threshold for defining hyperglycemia. However, data demonstrate that the majority of individuals without diabetes exceed this threshold every day. Moreover, people with diabetes spend >60% of their day above this threshold, which suggests that 140 mg/dL (7.8 mmol/L) is too low of a threshold for measuring hyperglycemia in individuals with diabetes. Current clinical guidelines for people with diabetes indicate that peak prandial glucose should not exceed 180 mg/dL (10.0 mmol/L). As such, the Steering Committee identified 180 mg/dL (10.0 mmol/L) as the initial threshold defining elevated glucose. […]

Very elevated glucose is defined as a glucose concentration >250 mg/dL (13.9 mmol/L). Evidence examining the impact of hyperglycemia does not examine the incremental effects of increasing blood glucose. However, blood glucose values exceeding 250 mg/dL (13.9 mmol/L) increase the risk for DKA (58), and HbA1c readings at that level have been associated with a high likelihood of complications.”

“An individual whose blood glucose levels rarely extend beyond the thresholds defined for hypo- and hyperglycemia is less likely to be subject to the short-term or long-term effects experienced by those with frequent excursions beyond one or both thresholds. It is also evident that if the intent of a given intervention is to safely manage blood glucose but the intervention does not reliably maintain blood glucose within safe levels, then the intervention should not be considered effective.

The time in range outcome is distinguished from traditional HbA1c testing in several ways (4,59). Time in range captures fluctuations in glucose levels continuously, whereas HbA1c testing is done at static points in time, usually months apart (60). Furthermore, time in range is more specific and sensitive than traditional HbA1c testing; for example, a treatment that addresses acute instances of hypo- or hyperglycemia may be detected in a time in range assessment but not necessarily in an HbA1c assessment. As a percentage, time in range is also more likely to be comparable across patients than HbA1c values, which are more likely to have patient-specific variations in significance (61). Finally, time in range may be more likely than HbA1c levels to correlate with PROs, such as quality of life, because the outcome is more representative of the whole patient experience (62). Table 3 illustrates how the concept of time in range differs from current HbA1c testing. […] [V]ariation in what is considered “normal” glucose fluctuations across populations, as well as what is realistically achievable for people with type 1 diabetes, must be taken into account so as not to make the target range definition too restrictive.”

“The Steering Committee defines time in range for individuals with type 1 diabetes as the following:

  • Percentage of readings in the range of 70–180 mg/dL (3.9–10.0 mmol/L) per unit of time

The Steering Committee considered it important to keep the time in range definition wide in order to accommodate variations across the population with type 1 diabetes — including different age-groups — but limited enough to preclude the possibility of negative outcomes. The upper and lower bounds of the time in range definition are consistent with the definitions for hypo- and hyperglycemia defined above. For individuals without type 1 diabetes, 70–140 mg/dL (3.9–7.8 mmol/L) represents a normal glycemic range (66). However, spending most of the day in this range is not generally achievable for people with type 1 diabetes […] To date, there is limited research correlating time in range with positive short-term and long-term type 1 diabetes outcomes, as opposed to the extensive research demonstrating the negative consequences of excursions into hyper- or hypoglycemia. More substantial evidence demonstrating a correlation or a direct causative relationship between time in range for patients with type 1 diabetes and positive health outcomes is needed.”

“DKA is often associated with hyperglycemia. In most cases, in an individual with diabetes, the cause of hyperglycemia is also the cause of DKA, although the two conditions are distinct. DKA develops when a lack of glucose in cells prompts the body to begin breaking down fatty acid reserves. This increases the levels of ketones in the body (ketosis) and causes a drop in blood pH (acidosis). At its most severe, DKA can cause cerebral edema, acute respiratory distress, thromboembolism, coma, and death (69,70). […] Although the current definition for DKA includes a list of multiple criteria that must be met, not all information currently included in the accepted definition is consistently gathered or required to diagnose DKA. The Steering Committee defines DKA in individuals with type 1 diabetes in a clinical setting as the following:

  • Elevated serum or urine ketones (greater than the upper limit of the normal range), and

  • Serum bicarbonate <15 mmol/L or blood pH <7.3

Given the seriousness of DKA, it is unnecessary to stratify DKA into different levels or categories, as the presence of DKA—regardless of the differences observed in the separate biochemical tests—should always be considered serious. In individuals with known diabetes, plasma glucose values are not necessary to diagnose DKA. Further, new therapeutic agents, specifically sodium–glucose cotransporter 2 inhibitors, have been linked to euglycemic DKA, or DKA with blood glucose values <250 mg/dL (13.9 mmol/L).”

“In guidance released in 2009 (72), the U.S. Food and Drug Administration (FDA) defined PROs as “any report of the status of a patient’s health condition that comes directly from the patient, without interpretation of the patient’s response by a clinician or anyone else.” In the same document, the FDA clearly acknowledged the importance of PROs, advising that they be used to gather information that is “best known by the patient or best measured from the patient perspective.”

Measuring and using PROs is increasingly seen as essential to evaluating care from a patient-centered perspective […] Given that type 1 diabetes is a chronic condition primarily treated on an outpatient basis, much of what people with type 1 diabetes experience is not captured through standard clinical measurement. Measures that capture PROs can fill these important information gaps. […] The use of validated PROs in type 1 diabetes clinical research is not currently widespread, and challenges to effectively measuring some PROs, such as quality of life, continue to confront researchers and developers.”

February 20, 2018 Posted by | Cardiology, Diabetes, Medicine, Neurology, Ophthalmology, Studies | Leave a comment

Prevention of Late-Life Depression (II)

Some more observations from the book:

In contrast to depression in childhood and youth when genetic and developmental vulnerabilities play a significant role in the development of depression, the development of late-life depression is largely attributed to its interactions with acquired factors, especially medical illness [17, 18]. An analysis of the WHO World Health Survey indicated that the prevalence of depression among medical patients ranged from 9.3 to 23.0 %, significantly higher than that in individuals without medical conditions [19]. Wells et al. [20] found in the Epidemiologic Catchment Area Study that the risk of developing lifetime psychiatric disorders among individuals with at least one medical condition was 27.9 % higher than among those without medical conditions. […] Depression and disability mutually reinforce the risk of each other, and adversely affect disease progression and prognosis [21, 25]. […] disability caused by medical conditions serves as a risk factor for depression [26]. When people lose their normal sensory, motor, cognitive, social, or executive functions, especially in a short period of time, they can become very frustrated or depressed. Inability to perform daily tasks as before decreases self-esteem, reduces independence, increases the level of psychological stress, and creates a sense of hopelessness. On the other hand, depression increases the risk for disability. Negative interpretation, attention bias, and learned hopelessness of depressed persons may increase risky health behaviors that exacerbate physical disorders or disability. Meanwhile, depression-related cognitive impairment also affects role performance and leads to functional disability [25]. For example, Egede [27] found in the 1999 National Health Interview Survey that the risk of having functional disability among patients with the comorbidity of diabetes and depression were approximately 2.5–5 times higher than those with either depression or diabetes alone. […]  A leading cause of disability among medical patients is pain and pain-related fears […] Although a large proportion of pain complaints can be attributed to physiological changes from physical disorders, psychological factors (e.g., attention, interpretation, and coping skills) play an important role in perception of pain […] Bair et al. [31] indicated in a literature review that the prevalence of pain was higher among depressed patients than non-depressed patients, and the prevalence of major depression was also higher among pain patients comparing to those without pain complaints.”

Alcohol use has more serious adverse health effects on older adults than other age groups, since aging-related physiological changes (e.g. reduced liver detoxification and renal clearance) affect alcohol metabolism, increase the blood concentration of alcohol, and magnify negative consequences. More importantly, alcohol interacts with a variety of frequently prescribed medications potentially influencing both treatment and adverse effects. […] Due to age-related changes in pharmacokinetics and pharmacodynamics, older adults are a vulnerable population to […] adverse drug effects. […] Adverse drug events are frequently due to failure to adjust dosage or to account for drug–drug interactions in older adults [64]. […] Loneliness […] is considered as an independent risk factor for depression [46, 47], and has been demonstrated to be associated with low physical activity, increased cardiovascular risks, hyperactivity of the hypothalamic-pituitary-adrenal axis, and activation of immune response [for details, see Cacioppo & Patrick’s book on these topics – US] […] Hopelessness is a key concept of major depression [54], and also an independent risk factor of suicidal ideation […] Hopelessness reduces expectations for the future, and negatively affects judgment for making medical and behavioral decisions, including non-adherence to medical regimens or engaging in unhealthy behaviors.”

Co-occurring depression and medical conditions are associated with more functional impairment and mortality than expected from the severity of the medical condition alone. For example, depression accompanying diabetes confers increased functional impairment [27], complications of diabetes [65, 66], and mortality [6771]. Frasure-Smith and colleagues highlighted the prognostic importance of depression among persons who had sustained a myocardial infarction (MI), finding that depression was a significant predictor of mortality at both 6 and 18 months post MI [72, 73]. Subsequent follow-up studies have borne out the increased risk conferred by depression on the mortality of patients with cardiovascular disease [10, 74, 75]. Over the course of a 2-year follow-up interval, depression contributed as much to mortality as did myocardial infarction or diabetes, with the population attributable fraction of mortality due to depression approximately 13 % (similar to the attributable risk associated with heart attack at 11 % and diabetes at 9 %) [76]. […] Although the bidirectional relationship between physical disorders and depression has been well known, there are still relatively few randomized controlled trials on preventing depression among medically ill patients. […] Rates of attrition [in post-stroke depression prevention trials has been observed to be] high […] Stroke, acute coronary syndrome, cancer, and other conditions impose a variety of treatment burdens on patients so that additional interventions without direct or immediate clinical effects may not be acceptable [95]. So even with good participation rates, lack of adherence to the intervention might limit effects.”

Late-life depression (LLD) is a heterogeneous disease, with multiple risk factors, etiologies, and clinical features. It has been recognized for many years that there is a significant relationship between the presence of depression and cerebrovascular disease in older adults [1, 2]. This subtype of LLD was eventually termed “vascular depression.” […] There have been a multitude of studies associating white matter abnormalities with depression in older adults using MRI technology to visualize lesions, or what appear as hyperintensities in the white matter on T2-weighted scans. A systematic review concluded that white matter hyperintensities (WMH) are more common and severe among older adults with depression compared to their non-depressed peers [9]. […] WMHs are associated with older age [13] and cerebrovascular risk factors, including diabetes, heart disease, and hypertension [14–17]. White matter severity and extent of WMH volume has been related to the severity of depression in late life [18, 19]. For example, among 639 older, community-dwelling adults, white matter lesion (WML) severity was found to predict depressive episodes and symptoms over a 3-year period [19]. […] Another way of investigating white matter integrity is with diffusion tensor imaging (DTI), which measures the diffusion of water in tissues and allows for indirect evidence of the microstructure of white matter, most commonly represented as fractional anisotropy (FA) and mean diffusivity (MD). DTI may be more sensitive to white matter pathology than is quantification of WMH […] A number of studies have found lower FA in widespread regions among individuals with LLD relative to controls [34, 36, 37]. […] lower FA has been associated with poorer performance on measures of cognitive functioning among patients with LLD [35, 38–40] and with measures of cerebrovascular risk severity. […] It is important to recognize that FA reflects the organization of fiber tracts, including fiber density, axonal diameter, or myelination in white matter. Thus, lower FA can result from multiple pathophysiological sources [42, 43]. […] Together, the aforementioned studies provide support for the vascular depression hypothesis. They demonstrate that white matter integrity is reduced in patients with LLD relative to controls, is somewhat specific to regions important for cognitive and emotional functioning, and is associated with cognitive functioning and depression severity. […] There is now a wealth of evidence to support the association between vascular pathology and depression in older age. While the etiology of depression in older age is multifactorial, from the epidemiological, neuroimaging, behavioral, and genetic evidence available, we can conclude that vascular depression represents one important subtype of LLD. The mechanisms underlying the relationship between vascular pathology and depression are likely multifactorial, and may include disrupted connections between key neural regions, reduced perfusion of blood to key brain regions integral to affective and cognitive processing, and inflammatory processes.”

Cognitive changes associated with depression have been the focus of research for decades. Results have been inconsistent, likely as a result of methodological differences in how depression is diagnosed and cognitive functioning measured, as well as the effects of potential subtypes and the severity of depression […], though deficits in executive functioning, learning and memory, and attention have been associated with depression in most studies [75]. In older adults, additional confounding factors include the potential presence of primary degenerative disorders, such as Alzheimer’s disease, which can pose a challenge to differential diagnosis in its early stages. […] LLD with cognitive dysfunction has been shown to result in greater disability than depressive symptoms alone [6], and MCI [mild cognitive impairment, US] with co-occurring LLD has been shown to double the risk of developing Alzheimer’s disease (AD) compared to MCI alone [86]. The conversion from MCI to AD also appears to occur earlier in patients with cooccurring depressive symptoms, as demonstrated by Modrego & Ferrandez [86] in their prospective cohort study of 114 outpatients diagnosed with amnestic MCI. […] Given accruing evidence for abnormal functioning of a number of cortical and subcortical networks in geriatric depression, of particular interest is whether these abnormalities are a reflection of the actively depressed state, or whether they may persist following successful resolution of symptoms. To date, studies have investigated this question through either longitudinal investigation of adults with geriatric depression, or comparison of depressed elders who are actively depressed versus those who have achieved symptom remission. Of encouragement, successful treatment has been reliably associated with normalization of some aspects of disrupted network functioning. For example, successful antidepressant treatment is associated with reduction of the elevated cerebral glucose metabolism observed during depressed states (e.g., [71–74]), with greater symptom reduction associated with greater metabolic change […] Taken together, these studies suggest that although a subset of the functional abnormalities observed during the LLD state may resolve with successful treatment, other abnormalities persist and may be tied to damage to the structural connectivity in important affective and cognitive networks. […] studies suggest a chronic decrement in cognitive functioning associated with LLD that is not adequately addressed through improvement of depressive symptoms alone.”

A review of the literature on evidence-based treatments for LLD found that about 50 % of patients improved on antidepressants, but that the number needed to treat (NNT) was quite high (NNT = 8, [139]) and placebo effects were significant [140]. Additionally, no difference was demonstrated in the effectiveness of one antidepressant drug class over another […], and in one-third of patients, depression was resistant to monotherapy [140]. The addition of medications or switching within or between drug classes appears to result in improved treatment response for these patients [140, 141]. A meta-analysis of patient-level variables demonstrated that duration of depressive symptoms and baseline depression severity significantly predicts response to antidepressant treatment in LLD, with chronically depressed older patients with moderate-to-severe symptoms at baseline experiencing more improvement in symptoms than mildly and acutely depressed patients [142]. Pharmacological treatment response appears to range from incomplete to poor in LLD with co-occurring cognitive impairment.”

“[C]ompared to other formulations of prevention, such as primary, secondary, or tertiary — in which interventions are targeted at the level of disease/stage of disease — the IOM conceptual framework involves interventions that are targeted at the level of risk in the population [2]. […] [S]elective prevention studies have an important “numbers” advantage — similar to that of indicated prevention trials: the relatively high incidence of depression among persons with key risk markers enables investigator to test interventions with strong statistical power, even with somewhat modest sample sizes. This fact was illustrated by Schoevers and colleagues [3], in which the authors were able to account for nearly 50 % of total risk of late-life depression with consideration of only a handful of factors. Indeed, research, largely generated by groups in the Netherlands and the USA, has identified that selective prevention may be one of the most efficient approaches to late-life depression prevention, as they have estimated that targeting persons at high risk for depression — based on risk markers such as medical comorbidity, low social support, or physical/functional disability — can yield theoretical numbers needed to treat (NNTs) of approximately 5–7 in primary care settings [4–7]. […] compared to the findings from selective prevention trials targeting older persons with general health/medical problems, […] trials targeting older persons based on sociodemographic risk factors have been more mixed and did not reveal as consistent a pattern of benefits for selective prevention of depression.”

Few of the studies in the existing literature that involve interventions to prevent depression and/or reduce depressive symptoms in older populations have included economic evaluations [13]. The identification of cost-effective interventions to provide to groups at high risk for depression is an important public health goal, as such treatments may avert or reduce a significant amount of the disease burden. […] A study by Katon and colleagues [8] showed that elderly patients with either subsyndromal or major depression had significantly higher medical costs during the previous 6 months than those without depression; total healthcare costs were $1,045 to $1,700 greater, and total outpatient/ambulatory costs ranged from being $763 to $979 more, on average. Depressed patients had greater usage of health resources in every category of care examined, including those that are not mental health-related, such as emergency department visits. No difference in excess costs was found between patients with a DSM-IV depressive disorder and those with depressive symptoms only, however, as mean total costs were 51 % higher in the subthreshold depression group (95 % CI = 1.39–1.66) and 49 % higher in the MDD/dysthymia group (95 % CI = 1.28–1.72) than in the nondepressed group [8]. In a similar study, the usage of various types of health services by primary care patients in the Netherlands was assessed, and average costs were determined to be 1,403 more in depressed individuals versus control patients [21]. Study investigators once again observed that patients with depression had greater utilization of both non-mental and mental healthcare services than controls.”

“In order for routine depression screening in the elderly to be cost-effective […] appropriate follow-up measures must be taken with those who screen positive, including a diagnostic interview and/or referral to a mental health professional [this – the necessity/requirement of proper follow-up following screens in order for screening to be cost-effective – is incidentally a standard result in screening contexts, see also Juth & Munthe’s book – US] [23, 25]. For example, subsequent steps may include initiation of psychotherapy or antidepressant treatment. Thus, one reason that the USPSTF does not recommend screening for depression in settings where proper mental health resources do not exist is that the evidence suggests that outcomes are unlikely to improve without effective follow-up care […]  as per the USPSTF suggestion, Medicare will only cover the screening when the appropriate supports for proper diagnosis and treatment are available […] In order to determine which interventions to prevent and treat depression should be provided to those who screen positive for depressive symptoms and to high-risk populations in general, cost-effectiveness analyses must be completed for a variety of different treatments and preventive measures. […] questions remain regarding whether annual versus other intervals of screening are most cost-effective. With respect to preventive interventions, the evidence to date suggests that these are cost-effective in settings where those at the highest risk are targeted.”

February 19, 2018 Posted by | Books, Cardiology, Diabetes, Health Economics, Neurology, Pharmacology, Psychiatry, Psychology | Leave a comment

Peripheral Neuropathy (II)

Chapter 3 included a great new (…new to me, that is…) chemical formula which I can’t not share here: (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl)pyrrolo[1,2,3-de]-1,4-benzoxazin-6-yl]-1-naphthalenylmethanonemesylate. It’s a cannabinoid receptor agonist, the properties of which are briefly discussed in the book‘s chapter 3.

Anyway, some more observations from the book below:

Injuries affecting either the peripheral or the central nervous system (PNS, CNS) leads to neuropathic pain characterized by spontaneous pain and distortion or exaggeration of pain sensation. Peripheral nerve pathologies are considered generally easier to treat compared to those affecting the CNS, however peripheral neuropathies still remain a challenge to therapeutic treatment. […] Although first being thought as a disease of purely neuronal nature, several pre-clinical studies indicate that the mechanisms at the basis of the development and maintenance of neuropathic pain involve substantial contributions from the nonneuronal cells of both the PNS and CNS [22]. After peripheral nerve injury, microglia in the normal conditions (usually defined ‘‘resting’’ microglia) in the spinal dorsal horn proliferate and change their phenotype to an “activated” state through a series of cellular and molecular changes. Microglia shift their phenotype to the hypertrophic “activated” form following altered expression of several molecules including cell surface receptors, intracellular signalling molecules and diffusible factors. The activation process consists of distinct cellular functions aimed at repairing damaged neural cells and eliminating debris from the damaged area [23]. Damaged cells release chemo-attractant molecules that both increase the motility (i.e. chemo‐kinesis) and stimulate the migration (i.e. chemotaxis) of microglia, the combination of which recruits the microglia much closer to the damaged cells […] Once microglia become activated, they can exert both proinflammatory or anti-inflammatory/neuroprotective functions depending on the combination of the stimulation of several receptors and the expression of specific genes [31]. Thus, the activation of microglia following a peripheral injury can be considered as an adaptation to tissue stress and malfunction [32] that contribute to the development and subsequent maintenance of chronic pain [33, 34]. […] The signals responsible for neuron-microglia and/or astrocyte communication are being extensively investigated since they may represent new targets for chronic pain management.”

“In the past two decades a notable increase in the incidence of [upper extremity compression neuropathies] has occurred. […] it is mandatory to achieve a prompt diagnosis because they can produce important motor and sensory deficiencies that need to be treated before the development of complications, since, despite the capacity for regeneration bestowed on the peripheral nervous system, functions lost as a result of denervation are never fully restored. […] There are many different situations that may be a direct cause of nerve compression. Anatomically, nerves can be compressed when traversing fibro-osseous tunnels, passing between muscle layers, through traction as they cross joints or buckling during certain movements of the wrist and elbow. Other causes include trauma, direct pressure and space-occupying lesions at any level in the upper extremity. There are other situations that are not a direct cause of nerve compression, but may increase the risk and may predispose the nerve to be compressed specially when the soft tissues are swollen like synovitis, pregnancy, hypothyroidism, diabetes or alcoholism [1]. […] When nerve fibers undergo compression, the response depends on the force applied at the site and the duration. Acute, brief compression results in a focal conduction block as a result of local ischemia, being reversible if the duration of compression is transient. On the other hand, if the focal compression is prolonged, ischemic changes appear, followed by endoneurial edema and secondary perineurial thickening. These histological alterations will aggravate the changes in the microneural circulation and will increase the sensitivity of the neuron sheath to ischemia. If the compression continues, we will find focal demyelination, which typically results in a greater involvement of motor than sensory nerve fibers. […] As the duration of compression increases beyond several hours, more diffuse demyelination will appear […] This process begins at the distal end of compression or injury, a process termed wallerian degeneration. These neural changes may not appear at a uniform fashion among the whole neural sheath depending on the distribution of the compressive forces, causing mixed demyelinating and axonal injury resulting from a combination of mechanical distortion of the nerve, ischemic injury, and impaired axonal flow [2].”

Electrophysiologic testing is part of the evaluation [of compression neuropathies], but it never substitutes a complete history and a thorough physical examination. These tests can detect physiologic abnormalities in the course of motor and sensory axons. There are two main electrophysiologic tests: needle electromyography and nerve conduction […] The electromyography detects the voluntary or spontaneous generated electrical activity. The registry of this activity is made through the needle insertion, at rest and during muscular activity to assess duration, amplitude, configuration and recruitment after injury. […] Nerve conduction assesses for both sensory and motor nerves. This study consists in applying a voltage simulator to the skin over different points of the nerve in order to record the muscular action potential, analyzing the amplitude, duration, area, latency and conduction velocity. The amplitude indicates the number of available nerve fibers.”

There are three well-described entrapment syndromes involving the median nerve or its branches, namely pronator teres syndrome, anterior interosseous syndrome and carpal tunnel syndrome according to the level of entrapment. Each one of these syndromes presents with different clinical signs and symptoms, electrophysiologic results and requires different techniques for their release. […] [In pronator teres syndrome] [t]he onset is insidious and is suggested when the early sensory disturbances are greater on the thumb and index finger, mainly tingling, numbness and dysaesthesia in the median nerve distribution. Patients will also complain of increased pain in the proximal forearm and greater hand numbness with sustained power gripping or rotation […] Surgical decompression is the definitive treatment. […] [Anterior interosseous syndrome] presents principally as weakness of the index finger and thumb, and the patient may complain of diffuse pain in the proximal forearm, which may be exacerbated during exercise and diminished with rest. The vast majority of patients begin with pain in the upper arm, elbow and forearm, often preceding the motor symptoms. […] During physical exam, the patient will be unable to bend the tip of the thumb and tip of index finger. The typical symptom is the inability to form an “O” with the thumb and index finger. […] If the onset was spontaneous and there is no evident lesion on MRI, supportive care and corticosteroid injections with observation for 4 to 6 weeks is usually accepted management. The degree of recovery is unpredictable.”

“[Carpal tunnel syndrome] is the most frequently encountered compression neuropathy in the upper limb. It is a mechanical compression of the median nerve through the fixed space of the rigid carpal tunnel. The incidence in the United States has been estimated at 1 to 3 cases per 1,000 subjects per year, with a prevalence of 50 cases per 1,000 subjects per year. [10] It is more common in women than in men (2:1), perhaps because the carpal tunnel itself may be smaller in women than in men. The dominant hand is usually affected first and produces the most severe pain. It usually occurs in adults […] Abnormalities on electrophysiologic testing, in association with specific symptoms and signs, are considered the criterion standard for carpal tunnel syndrome diagnosis. Electrophysiologic testing also can provide an accurate assessment of how severe the damage to the nerve is, thereby directing management and providing objective criteria for the determination of prognosis. Carpal tunnel syndrome is usually divided into mild, moderate and severe. In general, patients with mild carpal tunnel syndrome have sensory abnormalities alone on electrophysiologic testing, and patients with sensory plus motor abnormalities have moderate carpal tunnel syndrome. However, any evidence of axonal loss is classified as severe carpal tunnel syndrome. […] No imaging studies are considered routine in the diagnosis of carpal tunnel syndrome. […] nonoperative treatment is based in splintage of the wrist in a neutral position for three weeks and steroid injections. This therapy has variable results, with a success rate up to 76% during one year, but with a recurrence rate as high as 94%. Non-operative treatment is indicated in patients with intermittent symptoms, initial stages and during pregnancy [17]. The only definitive treatment for carpal tunnel syndrome is surgical expansion of the carpal tunnel by transection of the transverse carpal ligament.”

Postural control can be defined as the control of the body’s position in space for the purposes of balance and orientation. Balance is the ability to maintain or return the body’s centre of gravity within the limits of stability that are determined by the base of support. Spatial orientation defines our natural ability to maintain our body orientation in relation to the surrounding environment, in static and dynamic conditions. The representation of the body’s static and dynamic geometry may be largely based on muscle proprioceptive inputs that continuously inform the central nervous system about the position of each part of the body in relation to the others. Posture is built up by the sum of several basic mechanisms. […] Postural balance is dependent upon integration of signals from the somatosensory, visual and vestibular systems, to generate motor responses, with cognitive demands that vary according to the task, the age of the individuals and their ability to balance. Descending postural commands are multivariate in nature, and the motion at each joint is affected uniquely by input from multiple sensors.
The proprioceptive system provides information on joint angles, changes in joint angles, joint position and muscle length and tension; while the tactile system is associated mainly with sensations of touch, pressure and vibration. Visual influence on postural control results from a complex synergy that receives multimodal inputs. Vestibular inputs tonically activate the anti-gravity leg muscles and, during dynamic tasks, vestibular information contributes to head stabilization to enable successful gaze control, providing a stable reference frame from which to generate postural responses. In order to assess instability or walking difficulty, it is essential to identify the affected movements and circumstances in which they occur (i.e. uneven surfaces, environmental light, activity) as well as any other associated clinical manifestation that could be related
to balance, postural control, motor control, muscular force, movement limitations or sensory deficiency. The clinical evaluation should include neurological examination; special care should be taken to identify visual and vestibular disorders, and to assess static and dynamic postural control and gait.”

Polyneuropathy modify the amount and the quality of the sensorial information that is necessary for motor control, with increased instability during both, upright stance and gait. Patients with peripheral neuropathy may have decreased stability while standing and when subjected to dynamic balance conditions. […] Balance and gait difficulties are the most frequently cited cause of falling […] Patients with polyneuropathy who have ankle weakness are more likely to experience multiple and injurious falls than are those without specific muscle weakness. […] During upright stance, compared to healthy subjects, recordings of the centre of pressure in patients with diabetic neuropathy have shown larger sway [95-96, 102], as well as increased oscillation […] Compared to healthy subjects, diabetic patients may have poorer balance during standing in diminished light compared to full light and no light conditions [105] […] compared to patients with diabetes but no peripheral neuropathy, patients with diabetic peripheral neuropathy are more likely to report an injury during walking or standing, which may be more frequent when walking on irregular surfaces [110]. Epidemiological surveys have established that a reduction of leg proprioception is a risk factor for falls in the elderly [111-112]. Symptoms and signs of peripheral neuropathy are frequently found during physical examination of older subjects. These clinical manifestations may be related to diabetes mellitus, alcoholism, nutritional deficiencies, autoimmune diseases, among other causes. In this group of patients, loss of plantar sensation may be an important contributor to the dynamic balance deficits and increased risk of falls [34, 109]. […] Apart from sensorymotor compromise, fear of falling may relate to restriction and avoidance of activities, which results in loss of strength especially in the lower extremities, and may also be predictive for future falls [117-119].”

“In patients with various forms of peripheral neuropathy, the use of a cane, ankle orthoses or touching a wall [has been shown to improve] spatial and temporal measures of gait regularity while walking under challenging conditions. Additional hand contact of external objects may reduce postural instability caused by a deficiency of one or more senses. […] Contact of the index finger with a stationary surface can greatly attenuate postural instability during upright stance, even when the level of force applied is far below that necessary to provide mechanical support [42]. […] haptic information about postural sway derived from contact with other parts of the body can also increase stability […] Studies evaluating preventive and treatment strategies through excercise [sic – US] that could improve balance in patients with polyneuropathy are scarce. However, evidence support that physical activity interventions that increase activity probably do not increase the risk of falling in patients with diabetic peripheral neuropathy, and in this group of patients, specific training may improve gait speed, balance, muscle strength and joint mobility.”

“Postherpetic neuralgia (PHN) is a form of refractory chronic neuralgia that […] currently lacks any effective prophylaxis. […] PHN has a variety of symptoms and significantly affects patient quality of life [3-12]. Various studies have statistically analyzed predictive factors for PHN [13-23], but neither obvious pathogenesis nor established treatment has been clarified or established. We designed and conducted a study on the premise that statistical identification of significant predictors for PHN would contribute to the establishment of an evidence-based medicine approach to the optimal treatment of PHN. […] Previous studies have shown that older age, female sex, presence of a prodrome, greater rash severity, and greater acute pain severity are predictors of increased PHN [14-18, 25]. Some other potential predictors (ophthalmic localization, presence of anxiety and depression, presence of allodynia, and serological/virological factors) have also been studied [14, 18]. […] The participants were 73 patients with herpes zoster who had been treated at the pain clinic of our hospital between January 2008 and June 2010. […] Multivariate ordered logistic regression analysis was performed to identify predictive factors for PHN. […] advanced age and deep pain at first visit were identified as predictive factors for PHN. DM [diabetes mellitus – US] and pain reduced by bathing should also be considered as potential predictors of PHN [24].”

February 14, 2018 Posted by | Books, Diabetes, Infectious disease, Medicine, Neurology | Leave a comment

Peripheral Neuropathy (I)

The objective of this book is to update health care professionals on recent advances in the pathogenesis, diagnosis and treatment of peripheral neuropathy. This work was written by a group of clinicians and scientists with large expertise in the field.

The book is not the first book about this topic I’ve read, so a lot of the stuff included was of course review – however it’s a quite decent text, and I decided to blog it in at least some detail anyway. It’s somewhat technical and it’s probably not a very good introduction to this topic if you know next to nothing about neurology – in that case I’m certain Said’s book (see the ‘not’-link above) is a better option.

I have added some observations from the first couple of chapters below. As InTech publications like these explicitly encourage people to share the ideas and observations included in these books, I shall probably cover the book in more detail than I otherwise would have.

“Within the developing world, infectious diseases [2-4] and trauma [5] are the most common sources of neuropathic pain syndromes. The developed world, in contrast, suffers more frequently from diabetic polyneuropathy (DPN) [6, 7], post herpetic neuralgia (PHN) from herpes zoster infections [8], and chemotherapy-induced peripheral neuropathy (CIPN) [9, 10]. There is relatively little epidemiological data regarding the prevalence of neuropathic pain within the general population, but a few estimates suggest it is around 7-8% [11, 12]. Despite the widespread occurrence of neuropathic pain, treatment options are limited and often ineffective […] Neuropathic pain can present as on-going or spontaneous discomfort that occurs in the absence of any observable stimulus or a painful hypersensitivity to temperature and touch. […] people with chronic pain have increased incidence of anxiety and depression and reduced scores in quantitative measures of health related quality of life [15]. Despite significant progress in chronic and neuropathic pain research, which has led to the discovery of several efficacious treatments in rodent models, pain management in humans remains ineffective and insufficient [16]. The lack of translational efficiency may be due to inadequate animal models that do not faithfully recapitulate human disease or from biological differences between rodents and humans […] In an attempt to increase the efficacy of medical treatment for neuropathic pain, clinicians and researchers have been moving away from an etiology based classification towards one that is mechanism based. It is current practice to diagnose a person who presents with neuropathic pain according to the underlying etiology and lesion topography [17]. However, this does not translate to effective patient care as these classification criteria do not suggest efficacious treatment. A more apt diagnosis might include a description of symptoms and the underlying pathophysiology associated with those symptoms.”

Neuropathic pain has been defined […] as “pain arising as the direct consequence of a lesion or disease affecting the somatosensory system” [18]. This is distinct from nociceptive pain – which signals tissue damage through an intact nervous system – in underlying pathophysiology, severity, and associated psychological comorbidities [13]. Individuals who suffer from neuropathic pain syndromes report pain of higher intensity and duration than individuals with non-neuropathic chronic pain and have significantly increased incidence of depression, anxiety, and sleep disorders [13, 19]. […] individuals with seemingly identical diseases who both develop neuropathic pain may experience distinct abnormal sensory phenotypes. This may include a loss of sensory perception in some modalities and increased activity in others. Often a reduction in the perception of vibration and light touch is coupled with positive sensory symptoms such as paresthesia, dysesthesia, and pain[20]. Pain may manifest as either spontaneous, with a burning or shock-like quality, or as a hypersensitivity to mechanical or thermal stimuli [21]. This hypersensitivity takes two forms: allodynia, pain that is evoked from a normally non-painful stimulus, and hyperalgesia, an exaggerated pain response from a moderately painful stimulus. […] Noxious stimuli are perceived by small diameter peripheral neurons whose free nerve endings are distributed throughout the body. These neurons are distinct from, although anatomically proximal to, the low threshold mechanoreceptors responsible for the perception of vibration and light touch.”

In addition to hypersensitivity, individuals with neuropathic pain frequently experience ongoing spontaneous pain as a major source of discomfort and distress. […] In healthy individuals, a quiescent neuron will only generate an action potential when presented with a stimulus of sufficient magnitude to cause membrane depolarization. Following nerve injury, however, significant changes in ion channel expression, distribution, and kinetics lead to disruption of the homeostatic electric potential of the membrane resulting in oscillations and burst firing. This manifests as spontaneous pain that has a shooting or burning quality […] There is reasonable evidence to suggest that individual ion channels contribute to specific neuropathic pain symptoms […] [this observation] provides an intriguing therapeutic possibility: unambiguous pharmacologic ion channel blockers to relieve individual sensory symptoms with minimal unintended effects allowing pain relief without global numbness. […] Central sensitization leads to painful hypersensitivity […] Functional and structural changes of dorsal horn circuitry lead to pain hypersensitivity that is maintained independent of peripheral sensitization [38]. This central sensitization provides a mechanistic explanation for the sensory abnormalities that occur in both acute and chronic pain states, such as the expansion of hypersensitivity beyond the innervation territory of a lesion site, repeated stimulation of a constant magnitude leading to an increasing pain response, and pain outlasting a peripheral stimulus [39-41]. In healthy individuals, acute pain triggers central sensitization, but homeostatic sensitivity returns following clearance of the initial insult. In some individuals who develop neuropathic pain, genotype and environmental factors contribute to maintenance of central sensitization leading to spontaneous pain, hyperalgesia, and allodynia. […] Similarly, facilitation also results in a lowered activation threshold in second order neurons”.

“Chronic pain conditions are associated with vast functional and structural changes of the brain, when compared to healthy controls, but it is currently unclear which comes first: does chronic pain cause distortions of brain circuitry and anatomy or do cerebral abnormalities trigger and/or maintain the perception of chronic pain? […] Brain abnormalities in chronic pain states include modification of brain activity patterns, localized decreases in gray matter volume, and circuitry rerouting [53]. […] Chronic pain conditions are associated with localized reduction in gray matter volume, and the topography of gray matter volume reduction is dictated, at least in part, by the particular pathology. […] These changes appear to represent a form of plasticity as they are reversible when pain is effectively managed [63, 67, 68].”

“By definition, neuropathic pain indicates direct pathology of the nervous system while nociceptive pain is an indication of real or potential tissue damage. Due to the distinction in pathophysiology, conventional treatments prescribed for nociceptive pain are not very effective in treating neuropathic pain and vice versa [78]. Therefore the first step towards meaningful pain relief is an accurate diagnosis. […] Treating neuropathic pain requires a multifaceted approach that aims to eliminate the underlying etiology, when possible, and manage the associated discomforts and emotional distress. Although in some cases it is possible to directly treat the cause of neuropathic pain, for example surgery to alleviate a constricted nerve, it is more likely that the primary cause is untreatable, as is the case with singular traumatic events such as stroke and spinal cord injury and diseases like diabetes. When this is the case, symptom management and pain reduction become the primary focus. Unfortunately, in most cases complete elimination of pain is not a feasible endpoint; a pain reduction of 30% is considered to be efficacious [21]. Additionally, many pharmacological treatments require careful titration and tapering to prevent adverse effects and toxicity. This process may take several weeks to months, and ultimately the drug may be ineffective, necessitating another trial with a different medication. It is therefore necessary that both doctor and patient begin treatment with realistic expectations and goals.”

First-line medications for the treatment of neuropathic pain are those that have proven efficacy in randomized clinical trials (RCTs) and are consistent with pooled clinical observations [81]. These include antidepressants, calcium channel ligands, and topical lidocaine [15]. Tricyclic antidepressants (TCAs) have demonstrated efficacy in treating neuropathic pain with positive results in RCTs for central post-stroke pain, PHN, painful diabetic and non-diabetic polyneuropathy, and post-mastectomy pain syndrome [82]. However they do not seem to be effective in treating painful HIV-neuropathy or CIPN [82]. Duloxetine and venlafaxine, two selective serotonin norepinephrine reuptake inhibitors (SSNRIs), have been found to be effective in DPN and both DPN and painful polyneuropathies, respectively [81]. […] Gabapentin and pregabalin have also demonstrated efficacy in several neuropathic pain conditions including DPN and PHN […] Topical lidocaine (5% patch or gel) has significantly reduced allodynia associated with PHN and other neuropathic pain syndromes in several RCTs [81, 82]. With no reported systemic adverse effects and mild skin irritation as the only concern, lidocaine is an appropriate choice for treating localized peripheral neuropathic pain. In the event that first line medications, alone or in combination, are not effective at achieving adequate pain relief, second line medications may be considered. These include opioid analgesics and tramadol, pharmaceuticals which have proven efficacy in RCTs but are associated with significant adverse effects that warrant cautious prescription [15]. Although opioid analgesics are effective pain relievers in several types of neuropathic pain [81, 82, 84], they are associated with misuse or abuse, hypogonadism, constipation, nausea, and immunological changes […] Careful consideration should be given when prescribing opiates to patients who have a personal or family history of drug or alcohol abuse […] Deep brain stimulation, a neurosurgical technique by which an implanted electrode delivers controlled electrical impulses to targeted brain regions, has demonstrated some efficacy in treating chronic pain but is not routinely employed due to a high risk-to-benefit ratio [91]. […] A major challenge in treating neuropathic pain is the heterogeneity of disease pathogenesis within an individual etiological classification. Patients with seemingly identical diseases may experience completely different neuropathic pain phenotypes […] One of the biggest barriers to successful management of neuropathic pain has been the lack of understanding in the underlying pathophysiology that produces a pain phenotype. To that end, significant progress has been made in basic science research.”

In diabetes mellitus, nerves and their supporting cells are subjected to prolonged hyperglycemia and metabolic disturbances and this culminates in reversible/irreversible nervous system dysfunction and damage, namely diabetic peripheral neuropathy (DPN). Due to the varying compositions and extents of neurological involvements, it is difficult to obtain accurate and thorough prevalence estimates of DPN, rendering this microvascular complication vastly underdiagnosed and undertreated [1-4]. According to American Diabetes Association, DPN occurs to 60-70% of diabetic individuals [5] and represents the leading cause of peripheral neuropathies among all cases [6, 7].”

A quick remark: This number seems really high to me. I won’t rule out that it’s accurate if you go with highly sensitive measures of neuropathy, but the number of patients who will experience significant clinical sequelae as a result of DPN is in my opinion likely to be significantly lower than that. On a peripherally related note, it should however on the other hand also be kept in mind that although diabetes-related neurological complications may display some clustering in patient groups – which will necessarily decrease the magnitude of the problem – no single test will ever completely rule out neurological complications in a diabetic; a patient with a negative Semmes-Weinstein monofilament test may still have autonomic neuropathy. So assessing the full disease burden in the context of diabetes-related neurological complications cannot be done using only a single instrument, and the full disease burden is likely to be higher than individual estimates encountered in the literature (unless a full neurological workup was done, which is unlikely to be the case). They do go into more detail about subgroups, clinical significance, etc. below, but I thought this observation was important to add early on in this part of the coverage.

Because diverse anatomic distributions and fiber types may be differentially affected in patients with diabetes, the disease manifestations, courses and pathologies of clinical and subclinical DPN are rather heterogeneous and encompass a broad spectrum […] Current consensus divides diabetes-associated somatic neuropathic syndromes into the focal/multifocal and diffuse/generalized neuropathies [6, 14]. The first category comprises a group of asymmetrical, acute-in-onset and self-limited single lesion(s) of nerve injury or impairment largely resulting from the increased vulnerability of diabetic nerves to mechanical insults (Carpal Tunnel Syndrome) […]. Such mononeuropathies occur idiopathically and only become a clinical problem in association with aging in 5-10% of those affected. Therefore, focal neuropathies are not extensively covered in this chapter [16]. The rest of the patients frequently develop diffuse neuropathies characterized by symmetrical distribution, insidious onset and chronic progression. In particular, a distal symmetrical sensorimotor polyneuropathy accounts for 90% of all DPN diagnoses in type 1 and type 2 diabetics and affects all types of peripheral sensory and motor fibers in a temporally non-uniform manner [6, 17].
Symptoms begin with prickling, tingling, numbness, paresthesia, dysesthesia and various qualities of pain associated with small sensory fibers at the very distal end (toes) of lower extremities [1, 18]. Presence of the above symptoms together with abnormal nociceptive response of epidermal C and A-δ fibers to pain/temperature (as revealed by clinical examination) constitute the diagnosis of small fiber sensory neuropathy, which produces both painful and insensate phenotypes [19]. Painful diabetic neuropathy is a prominent, distressing and chronic experience in at least 10-30% of DPN populations [20, 21]. Its occurrence does not necessarily correlate with impairment in electrophysiological or quantitative sensory testing (QST). […] Large myelinated sensory fibers that innervate the dermis, such as Aβ, also become involved later on, leading to impaired proprioception, vibration and tactile detection, and mechanical hypoalgesia [19]. Following this “stocking-glove”, length-dependent and dying-back evolvement, neurodegeneration gradually proceeds to proximal muscle sensory and motor nerves. Its presence manifests in neurological testings as reduced nerve impulse conductions, diminished ankle tendon reflex, unsteadiness and muscle weakness [1, 24].
Both the absence of protective sensory response and motor coordination predispose neuropathic foot to impaired wound healing and gangrenous ulceration — often ensued by limb amputation in severe and/or advanced cases […]. Although symptomatic motor deficits only appear in later stages of DPN [25], motor denervation and distal atrophy can increase the rate of fractures by causing repetitive minor trauma or falls [24, 28]. Other unusual but highly disabling late sequelae of DPN include limb ischemia and joint deformity [6]; the latter also being termed Charcot’s neuroarthropathy or Charcot’s joints [1]. In addition to significant morbidities, several separate cohort studies provided evidence that DPN [29], diabetic foot ulcers [30] and increased toe vibration perception threshold (VPT) [31] are all independent risk factors for mortality.”

Unfortunately, current therapy for DPN is far from effective and at best only delays the onset and/or progression of the disease via tight glucose control […] Even with near normoglycemic control, a substantial proportion of patients still suffer the debilitating neurotoxic consequences of diabetes [34]. On the other hand, some with poor glucose control are spared from clinically evident signs and symptoms of neuropathy for a long time after diagnosis [37-39]. Thus, other etiological factors independent of hyperglycemia are likely to be involved in the development of DPN. Data from a number of prospective, observational studies suggested that older age, longer diabetes duration, genetic polymorphism, presence of cardiovascular disease markers, malnutrition, presence of other microvascular complications, alcohol and tobacco consumption, and higher constitutional indexes (e.g. weight and height) interact with diabetes and make for strong predictors of neurological decline [13, 32, 40-42]. Targeting some of these modifiable risk factors in addition to glycemia may improve the management of DPN. […] enormous efforts have been devoted to understanding and intervening with the molecular and biochemical processes linking the metabolic disturbances to sensorimotor deficits by studying diabetic animal models. In return, nearly 2,200 articles were published in PubMed central and at least 100 clinical trials were reported evaluating the efficacy of a number of pharmacological agents; the majority of them are designed to inhibit specific pathogenic mechanisms identified by these experimental approaches. Candidate agents have included aldose reductase inhibitors, AGE inhibitors, γ-linolenic acid, α-lipoic acid, vasodilators, nerve growth factor, protein kinase Cβ inhibitors, and vascular endothelial growth factor. Notwithstanding a fruitful of knowledge and promising results in animals, none has translated into definitive clinical success […] Based on the records published by National Institute of Neurological Disorders and Stroke (NINDS), a main source of DPN research, about 16,488 projects were funded at the expense of over $8 billion for the fiscal years of 2008 through 2012. Of these projects, an estimated 72,200 animals were used annually to understand basic physiology and disease pathology as well as to evaluate potential drugs [255]. As discussed above, however, the usefulness of these pharmaceutical agents developed through such a pipeline in preventing or reducing neuronal damage has been equivocal and usually halted at human trials due to toxicity, lack of efficacy or both […]. Clearly, the pharmacological translation from our decades of experimental modeling to clinical practice with regard to DPN has thus far not even [been] close to satisfactory.”

Whereas a majority of the drugs investigated during preclinical testing executed experimentally desired endpoints without revealing significant toxicity, more than half that entered clinical evaluation for treating DPN were withdrawn as a consequence of moderate to severe adverse events even at a much lower dose. Generally, using other species as surrogates for human population inherently encumbers the accurate prediction of toxic reactions for several reasons […] First of all, it is easy to dismiss drug-induced non-specific effects in animals – especially for laboratory rodents who do not share the same size, anatomy and physical activity with humans. […]  Second, some physiological and behavioral phenotypes observable in humans are impossible for animals to express. In this aspect, photosensitive skin rash and pain serve as two good examples of non-translatable side effects. Rodent skin differs from that of humans in that it has a thinner and hairier epidermis and distinct DNA repair abilities [260]. Therefore, most rodent stains used in diabetes modeling provide poor estimates for the probability of cutaneous hypersensitivity reactions to pharmacological treatments […] Another predicament is to assess pain in rodents. The reason for this is simple: these animals cannot tell us when, where or even whether they are experiencing pain […]. Since there is not any specific type of behavior to which painful reaction can be unequivocally associated, this often leads to underestimation of painful side effects during preclinical drug screening […] The third problem is that animals and humans have different pharmacokinetic and toxicological responses.”

“Genetic or chemical-induced diabetic rats or mice have been a major tool for preclinical pharmacological evaluation of potential DPN treatments. Yet, they do not faithfully reproduce many neuropathological manifestations in human diabetics. The difficulty of such begins with the fact that it is not possible to obtain in rodents a qualitative and quantitative expression of the clinical symptoms that are frequently presented in neuropathic diabetic patients, including spontaneous pain of different characteristics (e.g. prickling, tingling, burning, squeezing), paresthesia and numbness. As symptomatic changes constitute an important parameter of therapeutic outcome, this may well underlie the failure of some aforementioned drugs in clinical trials despite their good performance in experimental tests […] Development of nerve dysfunction in diabetic rodents also does not follow the common natural history of human DPN. […] Besides the lack of anatomical resemblance, the changes in disease severity are often missing in these models. […] importantly, foot ulcers that occur as a late complication to 15% of all individuals with diabetes [14] do not spontaneously develop in hyperglycemic rodents. Superimposed injury by experimental procedure in the foot pads of diabetic rats or mice may lend certain insight in the impaired wound healing in diabetes [278] but is not reflective of the chronic, accumulating pathological changes in diabetic feet of human counterparts. Another salient feature of human DPN that has not been described in animals is the predominant sensory and autonomic nerve damage versus minimal involvement of motor fibers [279]. This should elicit particular caution as the selective susceptibility is critical to our true understanding of the etiopathogenesis underlying distal sensorimotor polyneuropathy in diabetes. In addition to the lack of specificity, most animal models studied only cover a narrow spectrum of clinical DPN and have not successfully duplicated syndromes including proximal motor neuropathy and focal lesions [279].
Morphologically, fiber atrophy and axonal loss exist in STZ-rats and other diabetic rodents but are much milder compared to the marked degeneration and loss of myelinated and unmyelinated nerves readily observed in human specimens [280]. Of significant note, rodents are notoriously resistant to developing some of the histological hallmarks seen in diabetic patients, such as segmental and paranodal demyelination […] the simultaneous presence of degenerating and regenerating fibers that is characteristic of early DPN has not been clearly demonstrated in these animals [44]. Since such dynamic nerve degeneration/regeneration signifies an active state of nerve repair and is most likely to be amenable to therapeutic intervention, absence of this property makes rodent models a poor tool in both deciphering disease pathogenesis and designing treatment approaches […] With particular respect to neuroanatomy, a peripheral axon in humans can reach as long as one meter [296] whereas the maximal length of the axons innervating the hind limb is five centimeters in mice and twelve centimeters in rats. This short length makes it impossible to study in rodents the prominent length dependency and dying-back feature of peripheral nerve dysfunction that characterizes human DPN. […] For decades the cytoarchitecture of human islets was assumed to be just like those in rodents with a clear anatomical subdivision of β-cells and other cell types. By using confocal microscopy and multi-fluorescent labeling, it was finally uncovered that human islets have not only a substantially lower percentage of β-cell population, but also a mixed — rather than compartmentalized — organization of the different cell types [297]. This cellular arrangement was demonstrated to directly alter the functional performance of human islets as opposed to rodent islets. Although it is not known whether such profound disparities in cell composition and association also exist in the PNS, it might as well be anticipated considering the many sophisticated sensory and motor activities that are unique to humans. Considerable species difference also manifest at a molecular level. […] At least 80% of human genes have a counterpart in the mouse and rat genome. However, temporal and spatial expression of these genes can vary remarkably between humans and rodents, in terms of both extent and isoform specificity.”

“Ultimately, a fundamental problem associated with resorting to rodents in DPN research is to study a human disorder that takes decades to develop and progress in organisms with a maximum lifespan of 2-3 years. […] It is […] fair to say that a full clinical spectrum of the maturity-onset DPN likely requires a length of time exceeding the longevity of rodents to present and diabetic rodent models at best only help illustrate the very early aspects of the entire disease syndrome. Since none of the early pathogenetic pathways revealed in diabetic rodents will contribute to DPN in a quantitatively and temporally uniform fashion throughout the prolonged natural history of this disease, it is not surprising that a handful of inhibitors developed against these processes have not benefited patients with relatively long-standing neuropathy. As a matter of fact, any agents targeting single biochemical insults would be too little too late to treat a chronic neurological disorder with established nerve damage and pathogenetic heterogeneity […] It is important to point out that the present review does not argue against the ability of animal models to shed light on basic molecular, cellular and physiological processes that are shared among species. Undoubtedly, animal models of diabetes have provided abundant insights into the disease biology of DPN. Nevertheless, the lack of any meaningful advance in identifying a promising pharmacological target necessitates a reexamination of the validity of current DPN models as well as to offer a plausible alternative methodology to scientific approaches and disease intervention. […] we conclude that the fundamental species differences have led to misinterpretation of rodent data and overall failure of pharmacological investment. As more is being learned, it is becoming prevailing that DPN is a chronic, heterogeneous disease unlikely to benefit from targeting specific and early pathogenetic components revealed by animal studies.”

February 13, 2018 Posted by | Books, Diabetes, Genetics, Medicine, Neurology, Pharmacology | Leave a comment

Endocrinology (part 2 – pituitary)

Below I have added some observations from the second chapter of the book, which covers the pituitary gland.

“The pituitary gland is centrally located at the base of the brain in the sella turcica within the sphenoid bone. It is attached to the hypothalamus by the pituitary stalk and a fine vascular network. […] The pituitary measures around 13mm transversely, 9mm anteroposteriorly, and 6mm vertically and weighs approximately 100mg. It increases during pregnancy to almost twice its normal size, and it decreases in the elderly. *Magnetic resonance imaging (MRI) currently provides the optimal imaging of the pituitary gland. *Computed tomography (CT) scans may still be useful in demonstrating calcification in tumours […] and hyperostosis in association with meningiomas or evidence of bone destruction. […] T1– weighted images demonstrate cerebrospinal fluid (CSF) as dark grey and brain as much whiter. This imagining is useful for demonstrating anatomy clearly. […] On T1– weighted images, pituitary adenomas are of lower signal intensity than the remainder of the normal gland. […] The presence of microadenomas may be difficult to demonstrate.”

“Hypopituitarism refers to either partial or complete deficiency of anterior and/or posterior pituitary hormones and may be due to [primary] pituitary disease or to hypothalamic pathology which interferes with the hypothalamic control of the pituitary. Causes: *Pituitary tumours. *Parapituitary tumours […] *Radiotherapy […] *Pituitary infarction (apoplexy), Sheehan’s syndrome. *Infiltration of the pituitary gland […] *infection […] *Trauma […] *Subarachnoid haemorrhage. *Isolated hypothalamic-releasing hormone deficiency, e.g. Kallmann’s syndrome […] *Genetic causes [Let’s stop here: Point is, lots of things can cause pituitary problems…] […] The clinical features depend on the type and degree of hormonal deficits, and the rate of its development, in addition to whether there is intercurrent illness. In the majority of cases, the development of hypopituitarism follows a characteristic order, which secretion of GH [growth hormone, US], then gonadotrophins being affected first, followed by TSH [Thyroid-Stimulating Hormone, US] and ACTH [Adrenocorticotropic Hormone, US] secretion at a later stage. PRL [prolactin, US] deficiency is rare, except in Sheehan’s syndrome associated with failure of lactation. ADH [antidiuretic hormone, US] deficiency is virtually unheard of with pituitary adenomas but may be seen rarely with infiltrative disorders and trauma. The majority of the clinical features are similar to those occurring when there is target gland insufficiency. […] NB Houssay phenomenon. Amelioration of diabetes mellitus in patients with hypopituitarism due to reduction in counter-regulatory hormones. […] The aims of investigation of hypopituitarism are to biochemically assess the extent of pituitary hormone deficiency and also to elucidate the cause. […] Treatment involves adequate and appropriate hormone replacement […] and management of the underlying cause.”

“Apoplexy refers to infarction of the pituitary gland due to either haemorrhage or ischaemia. It occurs most commonly in patients with pituitary adenomas, usually macroadenomas […] It is a medical emergency, and rapid hydrocortisone replacement can be lifesaving. It may present with […] sudden onset headache, vomiting, meningism, visual disturbance, and cranial nerve palsy.”

“Anterior pituitary hormone replacement therapy is usually performed by replacing the target hormone rather than the pituitary or hypothalamic hormone that is actually deficient. The exceptions to this are GH replacement […] and when fertility is desired […] [In the context of thyroid hormone replacement:] In contrast to replacement in [primary] hypothyroidism, the measurement of TSH cannot be used to assess adequacy of replacment in TSH deficiency due to hypothalamo-pituitary disease. Therefore, monitoring of treatment in order to avoid under- and over-replacement should be via both clinical assessment and by measuring free thyroid hormone concentrations […] [In the context of sex hormone replacement:] Oestrogen/testosterone administration is the usual method of replacement, but gonadotrophin therapy is required if fertility is desired […] Patients with ACTH deficiency usually need glucocorticoid replacement only and do not require mineralcorticoids, in contrast to patients with Addison’s disease. […] Monitoring of replacement [is] important to avoid over-replacement which is associated with BP, elevated glucose and insulin, and reduced bone mineral density (BMD). Under-replacement leads to the non-specific symptoms, as seen in Addison’s disease […] Conventional replacement […] may overtreat patients with partial ACTH deficiency.”

“There is now a considerable amount of evidence that there are significant and specific consequences of GH deficiency (GDH) in adults and that many of these features improve with GH replacement therapy. […] It is important to differentiate between adult and childhood onset GDH. […] the commonest cause in childhood is an isolated variable deficiency of GH-releasing hormone (GHRH) which may resolve in adult life […] It is, therefore, important to retest patients with childhood onset GHD when linear growth is completed (50% recovery of this group). Adult onset. GHD usually occurs [secondarily] to a structural pituitary or parapituitary condition or due to the effects of surgical treatment or radiotherapy. Prevalence[:] *Adult onset GHD 1/10,000 *Adult GHD due to adult and childhood onset GHD 3/10,000. Benefits of GH replacement[:] *Improved QoL and psychological well-being. *Improved exercise capacity. *↑ lean body mass and reduced fat mass. *Prolonged GH replacement therapy (>12-24 months) has been shown to increase BMD, which would be expected to reduce fracture rate. *There are, as yet, no outcome studies in terms of cardiovascular mortality. However, GH replacement does lead to a reduction (~15%) in cholesterol. GH replacement also leads to improved ventricular function and ↑ left ventricular mass. […] All patients with GHD should be considered for GH replacement therapy. […] adverse effects experienced with GH replacement usually resolve with dose reduction […] GH treatment may be associated with impairment of insulin sensitivity, and therefore markers of glycemia should be monitored. […] Contraindications to GH replacement[:] *Active malignancy. *Benign intracranial hypertension. *Pre-proliferative/proliferative retinopathy in diabetes mellitus.”

“*Pituitary adenomas are the most common pituitary disease in adults and constitute 10-15% of primary brain tumours. […] *The incidence of clinically apparent pituitary disease is 1 in 10,000. *Pituitary carcinoma is very rare (<0.1% of all tumours) and is most commonly ACTH- or prolactin-secreting. […] *Microadenoma <1cm. *Macroadenoma >1cm. [In terms of the functional status of tumours, the break-down is as follows:] *Prolactinoma 35-40%. *Non-functioning 30-35%. Growth hormone (acromegaly) 10-15%. *ACTH adenoma (Cushing’s disease) 5-10% *TSH adenoma <5%. […] Pituitary disease is associated with an increased mortality, predominantly due to vascular disease. This may be due to oversecretion of GH or ACTH, hormone deficiencies or excessive replacement (e.g. of hydrocortisone).”

“*Prolactinomas are the commonest functioning pituitary tumour. […] Malignant prolactinomas are very rare […] [Clinical features of hyperprolactinaemia:] *Galactorrhoea (up to 90%♀, <10% ♂). *Disturbed gonadal function [menstrual disturbance, infertility, reduced libido, ED in ♂] […] Hyperprolactinaemia is associated with a long-term risk of BMD. […] Hypothyroidism and chronic renal failure are causes of hyperprolactinaemia. […] Antipsychotic agents are the most likely psychotrophic agents to cause hyperprolactinaemia. […] Macroadenomas are space-occupying tumours, often associated with bony erosion and/or cavernous sinus invasion. […] *Invasion of the cavernous sinus may lead to cranial nerve palsies. *Occasionally, very invasive tumours may erode bone and present with a CSF leak or [secondary] meningitis. […] Although microprolactinomas may expand in size without treatment, the vast majority do not. […] Macroprolactinomas, however, will continue to expand and lead to pressure effects. Definite treatment of the tumour is, therefore, necessary.”

“Dopamine agonist treatment […] leads to suppression of PRL in most patients [with prolactinoma], with [secondary] effects of normalization of gonadal function and termination of galactorrhoea. Tumour shrinkage occurs at a variable rate (from 24h to 6-12 months) and extent and must be carefully monitored. Continued shrinkage may occur for years. Slow chiasmal decompression will correct visual field defects in the majority of patients, and immediate surgical decompression is not necessary. […] Cabergoline is more effective in normalization of PRL in microprolactinoma […], with fewer side effects than bromocriptine. […] Tumour enlargement following initial shrinkage on treatment is usually due to non-compliance. […] Since the introduction of dopamine agonist treatment, transsphenoidal surgery is indicated only for patients who are resistant to, or intolerant of, dopamine agonist treatment. The cure rate for macroprolactinomas treated with surgery is poor (30%), and, therefore, drug treatment is first-line in tumours of all size. […] Standard pituitary irradiation leads to slow reduction (over years) of PRL in the majority of patients. […] Radiotherapy is not indicated in the management of patients with microprolactinomas. It is useful in the treatment of macroprolactinomas once the tumour has been shrunken away from the chiasm, only if the tumour is resistant.”

“Acromegaly is the clinical condition resulting from prolonged excessive GH and hence IGF-1 secretion in adults. GH secretion is characterized by blunting of pulsatile secretion and failure of GH to become undetectable during the 24h day, unlike normal controls. […] *Prevalence 40-86 cases/million population. Annual incidence of new cases in the UK is 4/million population. *Onset is insidious, and there is, therefore, often a considerable delay between onset of clinical features and diagnosis. Most cases are diagnosed at 40-60 years. […] Pituitary gigantism [is] [t]he clinical syndrome resulting from excess GH secretion in children prior to fusion of the epiphyses. […] growth velocity without premature pubertal manifestations should arouse suspicion of pituitary gigantism. […] Causes of acromegaly[:] *Pituitary adenoma (>99% of cases). Macroadenomas 60-80%, microadenomas 20-40%. […] The clinical features arise from the effects of excess GH/IGF-1, excess PRL in some (as there is co-secretion of PRL in a minority (30%) of tumours […] and the tumour mass. [Signs and symptoms:] * sweating -> 80% of patients. *Headaches […] *Tiredness and lethargy. *Joint pains. *Change in ring or shoe size. *Facial appearance. Coarse features […] enlarged nose […] prognathism […] interdental separation. […] Enlargement of hands and feet […] [Complications:] *Hypertension (40%). *Insulin resistance and impaired glucose tolerance (40%)/diabetes mellitus (20%). *Obstructive sleep apnea – due to soft tissue swelling […] Ischaemic heart disease and cerebrovascular disease.”

“Management of acromegaly[:] The management strategy depends on the individual patient and also on the tumour size. Lowering of GH is essential in all situations […] Transsphenoidal surgery […] is usually the first line for treatment in most centres. *Reported cure rates vary: 40-91% for microadenomas and 10-48% for macroadenomas, depending on surgical expertise. […] Using the definition of post-operative cure as mean GH <2.5 micrograms/L, the reported recurrence rate is low (6% at 5 years). Radiotherapy […] is usually reserved for patients following unsuccessful transsphenoidal surgery, only occasionally is it used as [primary] therapy. […] normalization of mean GH may take several years and, during this time, adjunctive medical treatment (usually with somatostatin analogues) is required. […] Radiotherapy can induce GH deficiency which may need GH therapy. […] Somatostatin analogues lead to suppresion of GH secretion in 20-60% of patients with acromegaly. […] some patients are partial responders, and although somatostatin analogues will lead to lowering of mean GH, they do not suppress to normal despite dose escalation. These drugs may be used as [primary] therapy where the tumour does not cause mass effects or in patients who have received surgery and/or radiotherapy who have elevated mean GH. […] Dopamine agonists […] lead to lowering of GH levels but, very rarely, lead to normalization of GH or IGF-1 (<30%). They may be helpful, particularly if there is coexistent secretion of PRL, and, in these cases, there may be significant tumour shrinkage. […] GH receptor antagonists [are] [i]ndicated for somatostatin non-responders.”

“Cushing’s syndrome is an illness resulting from excess cortisol secretion, which has a high mortality if left untreated. There are several causes of hypercortisolaemia which must be differentiated, and the commonest cause is iatrogenic (oral, inhaled, or topical steroids). […] ACTH-dependent Cushing’s must be differentiated from ACTH-independent disease (usually due to an adrenal adenoma, or, rarely, carcinoma […]). Once a diagnosis of ACTH-dependent disease has been established, it is important to differentiate between pituitary-dependent (Cushing’s disease) and ectopic secretion. […] [Cushing’s disease is rare;] annual incidence approximately 2/million. The vast majority of Cushing’s syndrome is due to a pituitary ACTH-secreting corticotroph microadenoma. […] The features of Cushing’s syndrome are progressive and may be present for several years prior to diagnosis. […] *Facial appearance – round plethoric complexion, acne and hirsutism, thinning of scalp hair. *Weight gain – truncal obesity, buffalo hump […] *Skin – thin and fragile […] easy bruising […] *Proximal muscle weakness. *Mood disturbance – labile, depression, insomnia, psychosis. *Menstrual disturbance. *Low libido and impotence. […] Associated features [include:] *Hypertension (>50%) due to mineralocorticoid effects of cortisol […] *Impaired glucose tolerance/diabetes mellitus (30%). *Osteopenia and osteoporosis […] *Vascular disease […] *Susceptibility to infections. […] Cushing’s is associated with a hypercoagulable state, with increased cardiovascular thrombotic risks. […] Hypercortisolism suppresses the thyroidal, gonadal, and GH axes, leading to lowered levels of TSH and thyroid hormones as well as reduced gonadotrophins, gonadal steroids, and GH.”

“Treatment of Cushing’s disease[:] Transsphenoidal surgery [is] the first-line option in most cases. […] Pituitary radiotherapy [is] usually administered as second-line treatment, following unsuccessful transsphenoidal surgery. […] Medical treatment [is] indicated during the preoperative preparation of patients or while awaiting radiotherapy to be effective or if surgery or radiotherapy are contraindicated. *Inhibitors of steroidogenesis: metyrapone is usually used first-line, but ketoconazole should be used as first-line in children […] Disadvantage of these agents inhibiting steroidogenesis is the need to increase the dose to maintain control, as ACTH secretion will increase as cortisol concentrations decrease. […] Successful treatment (surgery or radiotherapy) of Cushing’s disease leads to cortisol deficiency and, therefore, glucocorticoid replacement therapy is essential. […] *Untreated [Cushing’s] disease leads to an approximately 30-50% mortality at 5 years, owing to vascular disease and susceptibility to infections. *Treated Cushing’s syndrome has a good prognosis […] *Although the physical features and severe psychological disorders associated with Cushing’s improve or resolve within weeks or months of successful treatment, more subtle mood disturbance may persist for longer. Adults may also have impaired cognitive function. […] it is likely that there is an cardiovascular risk. *Osteoporosis will usually resolve in children but may not improve significantly in older patients. […] *Hypertension has been shown to resolve in 80% and diabetes mellitus in up to 70%. *Recent data suggests that mortality even with successful treatment of Cushing’s is increased significantly.”

“The term incidentaloma refers to an incidentally detected lesion that is unassociated with hormonal hyper- or hyposecretion and has a benign natural history. The increasingly frequent detection of these lesions with technological improvements and more widespread use of sophisticated imaging has led to a management challenge – which, if any, lesions need investigation and/or treatment, and what is the optimal follow-up strategy (if required at all)? […] *Imaging studies using MRI demonstrate pituitary microadenomas in approximately 10% of normal volunteers. […] Clinically significant pituitary tumours are present in about 1 in 1,000 patients. […] Incidentally detected microadenomas are very unlikely (<10%) to increase in size whereas larger incidentally detected meso- and macroadenomas are more likely (40-50%) to enlarge. Thus, conservative management in selected patients may be appropriate for microadenomas which are incidentally detected […]. Macroadenomas should be treated, if possible.”

“Non-functioning pituitary tumours […] are unassociated with clinical syndromes of anterior pituitary hormone excess. […] Non-functioning pituitary tumours (NFA) are the commonest pituitary macroadenoma. They represent around 28% of all pituitary tumours. […] 50% enlarge, if left untreated, at 5 years. […] Tumour behaviour is variable, with some tumours behaving in a very indolent, slow-growing manner and others invading the sphenoid and cavernous sinus. […] At diagnosis, approximately 50% of patients are gonadotrophin-deficient. […] The initial definitive management in virtually every case is surgical. This removes mass effects and may lead to some recovery of pituitary function in around 10%. […] The use of post-operative radiotherapy remains controversial. […] The regrowth rate at 10 years without radiotherapy approaches 45% […] administration of post-operative radiotherapy reduces this regrowth rate to <10%. […] however, there are sequelae to radiotherapy – with a significant long-term risk of hypopituitarism and a possible risk of visual deterioration and malignancy in the field of radiation. […] Unlike the case for GH- and PRL-secreting tumours, medical therapy for NFAs is usually unhelpful […] Gonadotrophinomas […] are tumours that arise from the gonadotroph cells of the pituitary gland and produce FSH, LH, or the α subunit. […] they are usually silent and unassociated with excess detectable secretion of LH and FSH […] [they] present in the same manner as other non-functioning pituitary tumours, with mass effects and hypopituitarism […] These tumours are managed as non-functioning tumours.”

“The posterior lobe of the pituitary gland arises from the forebrain and comprises up to 25% of the normal adult pituitary gland. It produces arginine vasopressin and oxytocin. […] Oxytoxin has no known role in ♂ […] In ♀, oxytoxin contracts the pregnant uterus and also causes breast duct smooth muscle contraction, leading to breast milk ejection during breastfeeding. […] However, oxytoxin deficiency has no known adverse effect on parturition or breastfeeding. […] Arginine vasopressin is the major determinant of renal water excretion and, therefore, fluid balance. It’s main action is to reduce free water clearance. […] Many substances modulate vasopressin secretion, including the catecholamines and opioids. *The main site of action of vasopressin is in the collecting duct and the thick ascending loop of Henle […] Diabetes Insipidus (DI) […] is defined as the passage of large volumes (>3L/24h) of dilute urine (osmolality <300mOsm/kg). [It may be] [d]ue to deficiency of circulating arginine vasopressin [or] [d]ue to renal resistance to vasopressin.” […lots of other causes as well – trauma, tumours, inflammation, infection, vascular, drugs, genetic conditions…]

Hyponatraemia […] Incidence *1-6% of hospital admissions Na<130mmol/L. *15-22% hospital admissions Na<135mmol/L. […] True clinically apparent hyponatraemia is associated with either excess water or salt deficiency. […] Features *Depend on the underlying cause and also on the rate of development of hyponatraemia. May develop once sodium reaches 115mmol/L or earlier if the fall is rapid. Level at 100mmol/L or less is life-threatening. *Features of excess water are mainly neurological because of brain injury […] They include confusion and headache, progressing to seizures and coma. […] SIADH [Syndrome of Inappropriate ADH, US] is a common cause of hyponatraemia. […] The elderly are more prone to SIADH, as they are unable to suppress ADH as efficiently […] ↑ risk of hyponatraemia with SSRIs. […] rapid overcorrection of hyponatraemia may cause central pontine myelinolysis (demyelination).”

“The hypothalamus releases hormones that act as releasing hormones at the anterior pituitary gland. […] The commonest syndrome to be associated with the hypothalamus is abnormal GnRH secretion, leading to reduced gonadotrophin secretion and hypogonadism. Common causes are stress, weight loss, and excessive exercise.”

January 14, 2018 Posted by | Books, Cancer/oncology, Cardiology, Diabetes, Epidemiology, Medicine, Nephrology, Neurology, Ophthalmology, Pharmacology | Leave a comment

A few diabetes papers of interest

i. Type 2 Diabetes in the Real World: The Elusive Nature of Glycemic Control.

“Despite U.S. Food and Drug Administration (FDA) approval of over 40 new treatment options for type 2 diabetes since 2005, the latest data from the National Health and Nutrition Examination Survey show that the proportion of patients achieving glycated hemoglobin (HbA1c) <7.0% (<53 mmol/mol) remains around 50%, with a negligible decline between the periods 2003–2006 and 2011–2014. The Healthcare Effectiveness Data and Information Set reports even more alarming rates, with only about 40% and 30% of patients achieving HbA1c <7.0% (<53 mmol/mol) in the commercially insured (HMO) and Medicaid populations, respectively, again with virtually no change over the past decade. A recent retrospective cohort study using a large U.S. claims database explored why clinical outcomes are not keeping pace with the availability of new treatment options. The study found that HbA1c reductions fell far short of those reported in randomized clinical trials (RCTs), with poor medication adherence emerging as the key driver behind the disconnect. In this Perspective, we examine the implications of these findings in conjunction with other data to highlight the discrepancy between RCT findings and the real world, all pointing toward the underrealized promise of FDA-approved therapies and the critical importance of medication adherence. While poor medication adherence is not a new issue, it has yet to be effectively addressed in clinical practice — often, we suspect, because it goes unrecognized. To support the busy health care professional, innovative approaches are sorely needed.”

“To better understand the differences between usual care and clinical trial HbA1c results, multivariate regression analysis assessed the relative contributions of key biobehavioral factors, including baseline patient characteristics, drug therapy, and medication adherence (21). Significantly, the key driver was poor medication adherence, accounting for 75% of the gap […]. Adherence was defined […] as the filling of one’s diabetes prescription often enough to cover ≥80% of the time one was recommended to be taking the medication (34). By this metric, proportion of days covered (PDC) ≥80%, only 29% of patients were adherent to GLP-1 RA treatment and 37% to DPP-4 inhibitor treatment. […] These data are consistent with previous real-world studies, which have demonstrated that poor medication adherence to both oral and injectable antidiabetes agents is very common (3537). For example, a retrospective analysis [of] adults initiating oral agents in the DPP-4 inhibitor (n = 61,399), sulfonylurea (n = 134,961), and thiazolidinedione (n = 42,012) classes found that adherence rates, as measured by PDC ≥80% at the 1-year mark after the initial prescription, were below 50% for all three classes, at 47.3%, 41.2%, and 36.7%, respectively (36). Rates dropped even lower at the 2-year follow-up (36)”

“Our current ability to assess adherence and persistence is based primarily on review of pharmacy records, which may underestimate the extent of the problem. For example, using the definition of adherence of the Centers for Medicare & Medicaid Services — PDC ≥80% — a patient could miss up to 20% of days covered and still be considered adherent. In retrospective studies of persistence, the permissible gap after the last expected refill date often extends up to 90 days (39,40). Thus, a patient may have a gap of up to 90 days and still be considered persistent.

Additionally, one must also consider the issue of primary nonadherence; adherence and persistence studies typically only include patients who have completed a first refill. A recent study of e-prescription data among 75,589 insured patients found that nearly one-third of new e-prescriptions for diabetes medications were never filled (41). Finally, none of these measures take into account if the patient is actually ingesting or injecting the medication after acquiring his or her refills.”

“Acknowledging and addressing the problem of poor medication adherence is pivotal because of the well-documented dire consequences: a greater likelihood of long-term complications, more frequent hospitalizations, higher health care costs, and elevated mortality rates (4245). In patients younger than 65, hospitalization risk in one study (n = 137,277) was found to be 30% at the lowest level of adherence to antidiabetes medications (1–19%) versus 13% at the highest adherence quintile (80–100%) […]. In patients over 65, a separate study (n = 123,235) found that all-cause hospitalization risk was 37.4% in adherent cohorts (PDC ≥80%) versus 56.2% in poorly adherent cohorts (PDC <20%) (45). […] Furthermore, for every 1,000 patients who increased adherence to their antidiabetes medications by just 1%, the total medical cost savings was estimated to be $65,464 over 3 years (45). […] “for reasons that are still unclear, the N.A. [North American] patient groups tend to have lower compliance and adherence compared to global rates during large cardiovascular studies” (46,47).”

“There are many potential contributors to poor medication adherence, including depressive affect, negative treatment perceptions, lack of patient-physician trust, complexity of the medication regimen, tolerability, and cost (48). […] A recent review of interventions addressing problematic medication adherence in type 2 diabetes found that few strategies have been shown consistently to have a marked positive impact, particularly with respect to HbA1c lowering, and no single intervention was identified that could be applied successfully to all patients with type 2 diabetes (53). Additional evidence indicates that improvements resulting from the few effective interventions, such as pharmacy-based counseling or nurse-managed home telemonitoring, often wane once the programs end (54,55). We suspect that the efficacy of behavioral interventions to address medication adherence will continue to be limited until there are more focused efforts to address three common and often unappreciated patient obstacles. First, taking diabetes medications is a burdensome and often difficult activity for many of our patients. Rather than just encouraging patients to do a better job of tolerating this burden, more work is needed to make the process easier and more convenient. […] Second, poor medication adherence often represents underlying attitudinal problems that may not be a strictly behavioral issue. Specifically, negative beliefs about prescribed medications are pervasive among patients, and behavioral interventions cannot be effective unless these beliefs are addressed directly (35). […] Third, the issue of access to medications remains a primary concern. A study by Kurlander et al. (51) found that patients selectively forgo medications because of cost; however, noncost factors, such as beliefs, satisfaction with medication-related information, and depression, are also influential.”

ii. Diabetes Research and Care Through the Ages. An overview article which might be of interest especially to people who’re not much familiar with the history of diabetes research and -treatment (a topic which is also very nicely covered in Tattersall’s book). Despite including a historical review of various topics, it also includes many observations about e.g. current (and future?) practice. Some random quotes:

“Arnoldo Cantani established a new strict level of treatment (9). He isolated his patients “under lock and key, and allowed them absolutely no food but lean meat and various fats. In the less severe cases, eggs, liver, and shell-fish were permitted. For drink the patients received water, plain or carbonated, and dilute alcohol for those accustomed to liquors, the total fluid intake being limited to one and one-half to two and one-half liters per day” (6).

Bernhard Naunyn encouraged a strict carbohydrate-free diet (6,10). He locked patients in their rooms for 5 months when necessary for “sugar-freedom” (6).” […let’s just say that treatment options have changed slightly over time – US]

“The characteristics of insulin preparations include the purity of the preparation, the concentration of insulin, the species of origin, and the time course of action (onset, peak, duration) (25). From the 1930s to the early 1950s, one of the major efforts made was to develop an insulin with extended action […]. Most preparations contained 40 (U-40) or 80 (U-80) units of insulin per mL, with U-10 and U-20 eliminated in the early 1940s. U-100 was introduced in 1973 and was meant to be a standard concentration, although U-500 had been available since the early 1950s for special circumstances. Preparations were either of mixed beef and pork origin, pure beef, or pure pork. There were progressive improvements in the purity of preparations as chemical techniques improved. Prior to 1972, conventional preparations contained 8% noninsulin proteins. […] In the early 1980s, “human” insulins were introduced (26). These were made either by recombinant DNA technology in bacteria (Escherichia coli) or yeast (Saccharomyces cerevisiae) or by enzymatic conversion of pork insulin to human insulin, since pork differed by only one amino acid from human insulin. The powerful nature of recombinant DNA technology also led to the development of insulin analogs designed for specific effects. These include rapid-acting insulin analogs and basal insulin analogs.”

“Until 1996, the only oral medications available were biguanides and sulfonylureas. Since that time, there has been an explosion of new classes of oral and parenteral preparations. […] The management of type 2 diabetes (T2D) has undergone rapid change with the introduction of several new classes of glucose-lowering therapies. […] the treatment guidelines are generally clear in the context of using metformin as the first oral medication for T2D and present a menu approach with respect to the second and third glucose-lowering medication (3032). In order to facilitate this decision, the guidelines list the characteristics of each medication including side effects and cost, and the health care provider is expected to make a choice that would be most suited for patient comorbidities and health care circumstances. This can be confusing and contributes to the clinical inertia characteristic of the usual management of T2D (33).”

“Perhaps the most frustrating barrier to optimizing diabetes management is the frequent occurrence of clinical inertia (whenever the health care provider does not initiate or intensify therapy appropriately and in a timely fashion when therapeutic goals are not reached). More broadly, the failure to advance therapy in an appropriate manner can be traced to physician behaviors, patient factors, or elements of the health care system. […] Despite clear evidence from multiple studies, health care providers fail to fully appreciate that T2D is a progressive disease. T2D is associated with ongoing β-cell failure and, as a consequence, we can safely predict that for the majority of patients, glycemic control will deteriorate with time despite metformin therapy (35). Continued observation and reinforcement of the current therapeutic regimen is not likely to be effective. As an example of real-life clinical inertia for patients with T2D on monotherapy metformin and an HbA1c of 7 to <8%, it took on the average 19 months before additional glucose-lowering therapy was introduced (36). The fear of hypoglycemia and weight gain are appropriate concerns for both patient and physician, but with newer therapies these undesirable effects are significantly diminished. In addition, health care providers must appreciate that achieving early and sustained glycemic control has been demonstrated to have long-term benefits […]. Clinicians have been schooled in the notion of a stepwise approach to therapy and are reluctant to initiate combination therapy early in the course of T2D, even if the combination intervention is formulated as a fixed-dose combination. […] monotherapy metformin failure rates with a starting HbA1c >7% are ∼20% per year (35). […] To summarize the current status of T2D at this time, it should be clearly emphasized that, first and foremost, T2D is characterized by a progressive deterioration of glycemic control. A stepwise medication introduction approach results in clinical inertia and frequently fails to meet long-term treatment goals. Early/initial combination therapies that are not associated with hypoglycemia and/or weight gain have been shown to be safe and effective. The added value of reducing CV outcomes with some of these newer medications should elevate them to a more prominent place in the treatment paradigm.”

iii. Use of Adjuvant Pharmacotherapy in Type 1 Diabetes: International Comparison of 49,996 Individuals in the Prospective Diabetes Follow-up and T1D Exchange Registries.

“The majority of those with type 1 diabetes (T1D) have suboptimal glycemic control (14); therefore, use of adjunctive pharmacotherapy to improve control has been of clinical interest. While noninsulin medications approved for type 2 diabetes have been reported in T1D research and clinical practice (5), little is known about their frequency of use. The T1D Exchange (T1DX) registry in the U.S. and the Prospective Diabetes Follow-up (DPV) registry in Germany and Austria are two large consortia of diabetes centers; thus, they provide a rich data set to address this question.

For the analysis, 49,996 pediatric and adult patients with diabetes duration ≥1 year and a registry update from 1 April 2015 to 1 July 2016 were included (19,298 individuals from 73 T1DX sites and 30,698 individuals from 354 DPV sites). Adjuvant medication use (metformin, glucagon-like peptide 1 [GLP-1] receptor agonists, dipeptidyl peptidase 4 [DPP-4] inhibitors, sodium–glucose cotransporter 2 [SGLT2] inhibitors, and other noninsulin diabetes medications including pramlintide) was extracted from participant medical records. […] Adjunctive agents, whose proposed benefits may include the ability to improve glycemic control, reduce insulin doses, promote weight loss, and suppress dysregulated postprandial glucagon secretion, have had little penetrance as part of the daily medical regimen of those in the registries studied. […] The use of any adjuvant medication was 5.4% in T1DX and 1.6% in DPV (P < 0.001). Metformin was the most commonly reported medication in both registries, with 3.5% in the T1DX and 1.3% in the DPV (P < 0.001). […] Use of adjuvant medication was associated with older age, higher BMI, and longer diabetes duration in both registries […] it is important to note that registry data did not capture the intent of adjuvant medications, which may have been to treat polycystic ovarian syndrome in women […here’s a relevant link, US].”

iv. Prevalence of and Risk Factors for Diabetic Peripheral Neuropathy in Youth With Type 1 and Type 2 Diabetes: SEARCH for Diabetes in Youth Study. I recently covered a closely related paper here (paper # 2) but the two papers cover different data sets so I decided it would be worth including this one in this post anyway. Some quotes:

“We previously reported results from a small pilot study comparing the prevalence of DPN in a subset of youth enrolled in the SEARCH for Diabetes in Youth (SEARCH) study and found that 8.5% of 329 youth with T1D (mean ± SD age 15.7 ± 4.3 years and diabetes duration 6.2 ± 0.9 years) and 25.7% of 70 youth with T2D (age 21.6 ± 4.1 years and diabetes duration 7.6 ± 1.8 years) had evidence of DPN (9). […this is the paper I previously covered here, US] Recently, we also reported the prevalence of microvascular and macrovascular complications in youth with T1D and T2D in the entire SEARCH cohort (10).

In the current study, we examined the cross-sectional and longitudinal risk factors for DPN. The aims were 1) to estimate prevalence of DPN in youth with T1D and T2D, overall and by age and diabetes duration, and 2) to identify risk factors (cross-sectional and longitudinal) associated with the presence of DPN in a multiethnic cohort of youth with diabetes enrolled in the SEARCH study.”

“The SEARCH Cohort Study enrolled 2,777 individuals. For this analysis, we excluded participants aged <10 years (n = 134), those with no antibody measures for etiological definition of diabetes (n = 440), and those with incomplete neuropathy assessment […] (n = 213), which reduced the analysis sample size to 1,992 […] There were 1,734 youth with T1D and 258 youth with T2D who participated in the SEARCH study and had complete data for the variables of interest. […] Seven percent of the participants with T1D and 22% of those with T2D had evidence of DPN.”

“Among youth with T1D, those with DPN were older (21 vs. 18 years, P < 0.0001), had a longer duration of diabetes (8.7 vs. 7.8 years, P < 0.0001), and had higher DBP (71 vs. 69 mmHg, P = 0.02), BMI (26 vs. 24 kg/m2, P < 0.001), and LDL-c levels (101 vs. 96 mg/dL, P = 0.01); higher triglycerides (85 vs. 74 mg/dL, P = 0.005); and lower HDL-c levels (51 vs. 55 mg/dL, P = 0.01) compared to those without DPN. The prevalence of DPN was 5% among nonsmokers vs. 10% among the current and former smokers (P = 0.001). […] Among youth with T2D, those with DPN were older (23 vs. 22 years, P = 0.01), had longer duration of diabetes (8.6 vs. 7.6 years; P = 0.002), and had lower HDL-c (40 vs. 43 mg/dL, P = 0.04) compared with those without DPN. The prevalence of DPN was higher among males than among females: 30% of males had DPN compared with 18% of females (P = 0.02). The prevalence of DPN was twofold higher in current smokers (33%) compared with nonsmokers (15%) and former smokers (17%) (P = 0.01). […] [T]he prevalence of DPN was further assessed by 5-year increment of diabetes duration in individuals with T1D or T2D […]. There was an approximately twofold increase in the prevalence of DPN with an increase in duration of diabetes from 5–10 years to >10 years for both the T1D group (5–13%) (P < 0.0001) and the T2D group (19–36%) (P = 0.02). […] in an unadjusted logistic regression model, youth with T2D were four times more likely to develop DPN compared with those with T1D, and though this association was attenuated, it remained significant independent of age, sex, height, and glycemic control (OR 2.99 [1.91; 4.67], P < 0.001)”.

“The prevalence estimates for DPN found in our study for youth with T2D are similar to those in the Australian cohort (8) but lower for youth with T1D than those reported in the Danish (7) and Australian (8) cohorts. The nationwide Danish Study Group for Diabetes in Childhood reported a prevalence of 62% among 339 adolescents and youth with T1D (age 12–27 years, duration 9–25 years, and HbA1c 9.7 ± 1.7%) using the vibration perception threshold to assess DPN (7). The higher prevalence in this cohort compared with ours (62 vs. 7%) could be due to the longer duration of diabetes (9–25 vs. 5–13 years) and reliance on a single measure of neuropathy (vibration perception threshold) as opposed to our use of the MNSI, which includes vibration as well as other indicators of neuropathy. In the Australian study, Eppens et al. (8) reported abnormalities in peripheral nerve function in 27% of the 1,433 adolescents with T1D (median age 15.7 years, median diabetes duration 6.8 years, and mean HbA1c 8.5%) and 21% of the 68 adolescents with T2D (median age 15.3 years, median diabetes duration 1.3 years, and mean HbA1c 7.3%) based on thermal and vibration perception threshold. These data are thus reminiscent of the persistent inconsistencies in the definition of DPN, which are reflected in the wide range of prevalence estimates being reported.”

“The alarming rise in rates of DPN for every 5-year increase in duration, coupled with poor glycemic control and dyslipidemia, in this cohort reinforces the need for clinicians rendering care to youth with diabetes to be vigilant in screening for DPN and identifying any risk factors that could potentially be modified to alter the course of the disease (2830). The modifiable risk factors that could be targeted in this young population include better glycemic control, treatment of dyslipidemia, and smoking cessation (29,30) […]. The sharp increase in rates of DPN over time is a reminder that DPN is one of the complications of diabetes that must be a part of the routine annual screening for youth with diabetes.”

v. Diabetes and Hypertension: A Position Statement by the American Diabetes Association.

“Hypertension is common among patients with diabetes, with the prevalence depending on type and duration of diabetes, age, sex, race/ethnicity, BMI, history of glycemic control, and the presence of kidney disease, among other factors (13). Furthermore, hypertension is a strong risk factor for atherosclerotic cardiovascular disease (ASCVD), heart failure, and microvascular complications. ASCVD — defined as acute coronary syndrome, myocardial infarction (MI), angina, coronary or other arterial revascularization, stroke, transient ischemic attack, or peripheral arterial disease presumed to be of atherosclerotic origin — is the leading cause of morbidity and mortality for individuals with diabetes and is the largest contributor to the direct and indirect costs of diabetes. Numerous studies have shown that antihypertensive therapy reduces ASCVD events, heart failure, and microvascular complications in people with diabetes (48). Large benefits are seen when multiple risk factors are addressed simultaneously (9). There is evidence that ASCVD morbidity and mortality have decreased for people with diabetes since 1990 (10,11) likely due in large part to improvements in blood pressure control (1214). This Position Statement is intended to update the assessment and treatment of hypertension among people with diabetes, including advances in care since the American Diabetes Association (ADA) last published a Position Statement on this topic in 2003 (3).”

“Hypertension is defined as a sustained blood pressure ≥140/90 mmHg. This definition is based on unambiguous data that levels above this threshold are strongly associated with ASCVD, death, disability, and microvascular complications (1,2,2427) and that antihypertensive treatment in populations with baseline blood pressure above this range reduces the risk of ASCVD events (46,28,29). The “sustained” aspect of the hypertension definition is important, as blood pressure has considerable normal variation. The criteria for diagnosing hypertension should be differentiated from blood pressure treatment targets.

Hypertension diagnosis and management can be complicated by two common conditions: masked hypertension and white-coat hypertension. Masked hypertension is defined as a normal blood pressure in the clinic or office (<140/90 mmHg) but an elevated home blood pressure of ≥135/85 mmHg (30); the lower home blood pressure threshold is based on outcome studies (31) demonstrating that lower home blood pressures correspond to higher office-based measurements. White-coat hypertension is elevated office blood pressure (≥140/90 mmHg) and normal (untreated) home blood pressure (<135/85 mmHg) (32). Identifying these conditions with home blood pressure monitoring can help prevent overtreatment of people with white-coat hypertension who are not at elevated risk of ASCVD and, in the case of masked hypertension, allow proper use of medications to reduce side effects during periods of normal pressure (33,34).”

“Diabetic autonomic neuropathy or volume depletion can cause orthostatic hypotension (35), which may be further exacerbated by antihypertensive medications. The definition of orthostatic hypotension is a decrease in systolic blood pressure of 20 mmHg or a decrease in diastolic blood pressure of 10 mmHg within 3 min of standing when compared with blood pressure from the sitting or supine position (36). Orthostatic hypotension is common in people with type 2 diabetes and hypertension and is associated with an increased risk of mortality and heart failure (37).

It is important to assess for symptoms of orthostatic hypotension to individualize blood pressure goals, select the most appropriate antihypertensive agents, and minimize adverse effects of antihypertensive therapy.”

“Taken together, […] meta-analyses consistently show that treating patients with baseline blood pressure ≥140 mmHg to targets <140 mmHg is beneficial, while more intensive targets may offer additional though probably less robust benefits. […] Overall, compared with people without diabetes, the relative benefits of antihypertensive treatment are similar, and absolute benefits may be greater (5,8,40). […] Multiple-drug therapy is often required to achieve blood pressure targets, particularly in the setting of diabetic kidney disease. However, the use of both ACE inhibitors and ARBs in combination is not recommended given the lack of added ASCVD benefit and increased rate of adverse events — namely, hyperkalemia, syncope, and acute kidney injury (7173). Titration of and/or addition of further blood pressure medications should be made in a timely fashion to overcome clinical inertia in achieving blood pressure targets. […] there is an absence of high-quality data available to guide blood pressure targets in type 1 diabetes. […] Of note, diastolic blood pressure, as opposed to systolic blood pressure, is a key variable predicting cardiovascular outcomes in people under age 50 years without diabetes and may be prioritized in younger adults (46,47). Though convincing data are lacking, younger adults with type 1 diabetes might more easily achieve intensive blood pressure levels and may derive substantial long-term benefit from tight blood pressure control.”

“Lifestyle management is an important component of hypertension treatment because it lowers blood pressure, enhances the effectiveness of some antihypertensive medications, promotes other aspects of metabolic and vascular health, and generally leads to few adverse effects. […] Lifestyle therapy consists of reducing excess body weight through caloric restriction, restricting sodium intake (<2,300 mg/day), increasing consumption of fruits and vegetables […] and low-fat dairy products […], avoiding excessive alcohol consumption […] (53), smoking cessation, reducing sedentary time (54), and increasing physical activity levels (55). These lifestyle strategies may also positively affect glycemic and lipid control and should be encouraged in those with even mildly elevated blood pressure.”

“Initial treatment for hypertension should include drug classes demonstrated to reduce cardiovascular events in patients with diabetes: ACE inhibitors (65,66), angiotensin receptor blockers (ARBs) (65,66), thiazide-like diuretics (67), or dihydropyridine CCBs (68). For patients with albuminuria (urine albumin-to-creatinine ratio [UACR] ≥30 mg/g creatinine), initial treatment should include an ACE inhibitor or ARB in order to reduce the risk of progressive kidney disease […]. In the absence of albuminuria, risk of progressive kidney disease is low, and ACE inhibitors and ARBs have not been found to afford superior cardioprotection when compared with other antihypertensive agents (69). β-Blockers may be used for the treatment of coronary disease or heart failure but have not been shown to reduce mortality as blood pressure–lowering agents in the absence of these conditions (5,70).”

vi. High Illicit Drug Abuse and Suicide in Organ Donors With Type 1 Diabetes.

“Organ donors with type 1 diabetes represent a unique population for research. Through a combination of immunological, metabolic, and physiological analyses, researchers utilizing such tissues seek to understand the etiopathogenic events that result in this disorder. The Network for Pancreatic Organ Donors with Diabetes (nPOD) program collects, processes, and distributes pancreata and disease-relevant tissues to investigators throughout the world for this purpose (1). Information is also available, through medical records of organ donors, related to causes of death and psychological factors, including drug use and suicide, that impact life with type 1 diabetes.

We reviewed the terminal hospitalization records for the first 100 organ donors with type 1 diabetes in the nPOD database, noting cause, circumstance, and mechanism of death; laboratory results; and history of illicit drug use. Donors were 45% female and 79% Caucasian. Mean age at time of death was 28 years (range 4–61) with mean disease duration of 16 years (range 0.25–52).”

“Documented suicide was found in 8% of the donors, with an average age at death of 21 years and average diabetes duration of 9 years. […] Similarly, a type 1 diabetes registry from the U.K. found that 6% of subjects’ deaths were attributed to suicide (2). […] Additionally, we observed a high rate of illicit substance abuse: 32% of donors reported or tested positive for illegal substances (excluding marijuana), and multidrug use was common. Cocaine was the most frequently abused substance. Alcohol use was reported in 35% of subjects, with marijuana use in 27%. By comparison, 16% of deaths in the U.K. study were deemed related to drug misuse (2).”

“We fully recognize the implicit biases of an organ donor–based population, which may not be […’may not be’ – well, I guess that’s one way to put it! – US] directly comparable to the general population. Nevertheless, the high rate of suicide and drug use should continue to spur our energy and resources toward caring for the emotional and psychological needs of those living with type 1 diabetes. The burden of type 1 diabetes extends far beyond checking blood glucose and administering insulin.”

January 10, 2018 Posted by | Cardiology, Diabetes, Epidemiology, Medicine, Nephrology, Neurology, Pharmacology, Psychiatry, Studies | Leave a comment