Skip to main content
Erschienen in: Reviews in Endocrine and Metabolic Disorders 4/2020

Open Access 17.03.2020

Hyperinsulinemic hypoglycemia in children and adolescents: Recent advances in understanding of pathophysiology and management

verfasst von: Maria Gϋemes, Sofia Asim Rahman, Ritika R. Kapoor, Sarah Flanagan, Jayne A. L. Houghton, Shivani Misra, Nick Oliver, Mehul Tulsidas Dattani, Pratik Shah

Erschienen in: Reviews in Endocrine and Metabolic Disorders | Ausgabe 4/2020

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Hyperinsulinemic hypoglycemia (HH) is characterized by unregulated insulin release, leading to persistently low blood glucose concentrations with lack of alternative fuels, which increases the risk of neurological damage in these patients. It is the most common cause of persistent and recurrent hypoglycemia in the neonatal period. HH may be primary, Congenital HH (CHH), when it is associated with variants in a number of genes implicated in pancreatic development and function. Alterations in fifteen genes have been recognized to date, being some of the most recently identified mutations in genes HK1, PGM1, PMM2, CACNA1D, FOXA2 and EIF2S3. Alternatively, HH can be secondary when associated with syndromes, intra-uterine growth restriction, maternal diabetes, birth asphyxia, following gastrointestinal surgery, amongst other causes. CHH can be histologically characterized into three groups: diffuse, focal or atypical. Diffuse and focal forms can be determined by scanning using fluorine-18 dihydroxyphenylalanine-positron emission tomography. Newer and improved isotopes are currently in development to provide increased diagnostic accuracy in identifying lesions and performing successful surgical resection with the ultimate aim of curing the condition. Rapid diagnostics and innovative methods of management, including a wider range of treatment options, have resulted in a reduction in co-morbidities associated with HH with improved quality of life and long-term outcomes. Potential future developments in the management of this condition as well as pathways to transition of the care of these highly vulnerable children into adulthood will also be discussed.
Hinweise

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

1 Introduction

Glucose is one of the principal energy substrates, providing half of the body’s total energy requirements. As the brain can neither synthesize nor store more than a few minutes supply of glucose, its function is solely dependent on maintenance of normal glucose concentrations in the circulation. An abnormally reduced concentration of glucose in the blood is referred to as hypoglycemia. It is a medical emergency and can lead to symptoms due to neuroglycopenia [1].
In healthy individuals, maintenance of a normal plasma glucose concentration relies on a tightly controlled balance between glucose production (dietary intake, glycogenolysis, gluconeogenesis) and its utilization by the tissues (glycolysis, glycogenesis, conversion to fatty acids). A normal endocrine system is essential for integrating and modulating substrate mobilization, interconversion, and utilization. In addition, the endocrine system interacts with metabolic pathways that rely critically on functionally intact enzymes. There are two types of metabolic hormones affecting blood glucose concentrations – an anabolic hormone (insulin), which decreases blood glucose, and several catabolic hormones (such as glucagon, cortisol and catecholamines) which increase blood glucose concentrations.
Hyperinsulinemic hypoglycemia (HH) is the commonest cause of persistent hypoglycemia in infants and children [2] and it can be transient –associated to risk factors- or permanent –linked to genetic mutations-. The risk of permanent brain injury in infants with HH continues to be as high as 25–50% due to delays in diagnosis and inadequate treatment. Despite advances in genetics, improved modes of investigation, novel management options and abrigding pediatric and adult follow-up in holistic multidisciplinary transition clinics, significant morbidity and mortality is still a major issue in children and young adults with HH [35].
The present review has been written using a comprehensive and up-to-date literature search on congenital hyperinsulinism/HH including the latest publications available in PubMed (last search in August 2019). It also incorporates clinical and laboratory experience from reference centers for the diagnosis and management of HH, as well as available data from on-going pharmaceutical trials.

2 normal blood glucose and hypoglycemia

2.1 Definition of normal blood glucose

Blood glucose concentrations of normal term neonates appropriate for gestational age may range between 1.4–6.2 mmol/l (25–112 mg/dl) during the first 72 h of life; however after that, healthy children and adults will maintain blood glucose concentrations between 3.5–5.5 mmol/l (63–99 mg/dl) [6]. It is difficult to numerically define hypoglycemia given that a single cut-off value cannot suit all individuals in every situation. Therefore operational thresholds are recommended which indicate that in any baby with clinical signs of hypoglycemia, blood glucose levels must be maintained over 2.6 mmol/l (47 mg/dl) except for suspected cases of hyperinsulinemic hypoglycemia in which 3.5 mmol/l (63 mg/dl) should be the cut-off point [7]. However, the Pediatric Endocrinology Society recommends that when a congenital disorder causing hypoglycemia is suspected in a neonate and when confirmed in older infants and children, the aim is to keep plasma glucose concentrations over 3.9 mmol/l (70 mg/dl) [7].

2.2 Causes of hypoglycemia

For hypoglycemia to occur, the rate of appearance of glucose into the plasma space must be less than its rate of utilization [8]. This can be due to defective glucose production, increased glucose utilization, or some combination of the two. Excessive glucose utilization due to hyperinsulinism (exogenous/endogenous) is one of the commonest causes of hypoglycemia. Hypoglycemia can also occur due to deficiencies of various counter regulatory hormones. The causes are collected in Table 1.
Table 1
Endocrine and metabolic causes of Hypoglycemia - Specific pathologies affecting main metabolic and endocrine pathways that can lead to hypoglycemia [5, 914, 61, 63]
Hyperinsulinism
Transient
Infant of diabetic mother
Perinatal asphyxia
Rhesus hemolytic disease
Intrauterine growth restriction
HNF4A/HNF1A
Congenital
ABCC8/ KCNJ11/ GCK/ GDH/ HADH/ HNF4A/ HNF1A/ UCP2/ SLC16A1/PMM2/HK1/PGM1/FOXA2/CACNA1D/EIF2S3
Others
Post-prandial hyperinsulinemic hypoglycemia
Insulinoma
Munchausen’s by proxy
Exercise induced hyperinsulinemic hypoglycemia
Hypoinsulinemic hypoglycemia
Activating AKT2 mutations
Counter-regulatory hormone deficiency
Growth hormone deficiency
Adrenal insufficiency
Fatty acid oxidation disorders
Medium chain acyl-CoA dehydrogenase deficiency
Long chain acyl-CoA dehydrogenase deficiency
Short chain acyl-CoA dehydrogenase deficiency
Defects in ketone body synthesis/ utilization
HMG CoA synthase deficiency
HMG CoA lyase deficiency
Carnitine deficiency (primary and secondary)
Carnitine palmitoyl transferase deficiency
(CPT 1 and 2), Carnitine deficiency
Gluconeogenic disorders
Fructose-1, 6-bisphosphatase deficiency,
Phosphoenolpyruvate carboxykinase (PEPCK) deficiency
Pyruvate carboxylase deficiency
Glycogen storage disorders
Glucose-6-phosphatase deficiency
Amylo 1–6 glucosidase deficiency
Glycogen synthase deficiency
Defects in glucose transport
GLUT 1/2/3 transporters defects
Other metabolic conditions
Galactosemia, Fructosemia, Tyrosinemia, Glutaric aciduria type 2, Maple syrup urine disease, Propionic academia
Adenosine kinase deficiency
Mitochondrial respiratory chain disease
Hereditary disorders caused by deficiency of specific enzymes involved in mobilization, interconversion, or utilization of metabolic substrates frequently are associated with hypoglycemia. These enzymatic defects may involve carbohydrate, amino acid, or fat metabolism and are individually rare; almost all are inherited as autosomal recessive traits [8].

3 HYPERINSULINEMIC hypoglycemia (HH)

HH is a condition caused by the upregulation of β-cell secretion of insulin producing a hypoglycemic state. Congenital hyperinsulinism (CHH) is the most common cause of transient or permanent hypoglycemia and could potentially be life threatening causing neurological damage. Hence it requires quick and effective treatment and management [8]. This disorder is rare and has an incidence of around 1:40,000 births in the general population [15]. CHH can occur due to genetic mutations and one of the most common causes are defects in the β-cell ATP-sensitive potassium (KATP) channels, known as channelopathies [8]. KATP channels are comprised of two subunits; the inward rectifying Kir6.2 channels and the sulphonylurea receptor-1, SUR-1, which are encoded for by the KCNJ11 (potassium voltage-gated channel subfamily J member 11) and ABCC8 (ATP-binding cassette transporter sub-family C member 8) genes, respectively [16]. Both these subunits are sensitive to the ADP/ATP nucleotide ratio and work together to promote cell depolarization and eventual insulin secretion. Mutations in the KCNJ11/ABCC8 genes are known to cause defects in biogenesis/trafficking of these subunits to the plasma membrane, thus causing HH.

4 Causes of HH

4.1 Transient forms of HH

Transient HH is a poorly defined term that refers to the group of patients in whom HH spontaneously resolves within a few days to approximately a week. However, the cohort includes children requiring medications up to 6 months of life and is usually negative for a known genetic etiology for HH [17]. It is associated with intra-uterine growth retardation, erythroblastosis fetalis, perinatal asphyxia, maternal diabetes mellitus (gestational or insulin dependent) and after the maternal administration of drugs such as sulphonylureas, and intravenous glucose infusions during labor [2]. Abnormal neurodevelopment is evident in one third of children with transient forms of HH associated with perinatal risk factors [5].

4.2 Permanent form of HH

A permanent form of HH, usually congenital (CHH), is where children continue to need medical treatment even after 6 months of age. Various genetic causes have been identified, however nearly 40–50% of children still remain genetically unidentified [17].

4.2.1 Molecular basis of CHH

To date, at least 15 genes have been identified to be accompanied with CHH, which include ABCC8, KCNJ11, GLUD1, GCK, HADH, SLC16A1, UCP2, HNF1A, HNF4A, HK1, PGM1, PMM2, FOXA2, CACNA1D and EIF2S3.
Various modes of inheritance are observed. For some patients specific clinical characteristics, such as the presence of hyperammonemia, can help guide molecular testing; however, for most of the genetic subgroups there is an overlap in phenotype and as such testing of all the known genes is often required.
KATP channel genes (ABCC8 and KCNJ11)
The pancreatic KATP channel is a key component of the insulin secretion pathway. Following glycolysis ATP binds to and closes the KATP channel causing membrane depolarization, opening of calcium channel and insulin exocytosis (Fig. 1). Loss of function variants in ABCC8 and KCNJ11 are the most common cause of HH accounting for 40–50% of cases [1719](OMIM #601820 and #256450). These variants exert their effects by 1) leading to a loss of KATP channels at the plasma membrane via effects on gene expression, protein synthesis, protein maturation, or membrane trafficking or 2) by impairing the ability of SUR1 to regulate channel activity by reducing or abolishing channel activation by MgADP and/or MgATP [20, 21]. Recessively inherited variants are the most common and usually result in medically unresponsive HH. Dominant variants in both genes have also been reported [2224]. The functionally more severe dominant variants cause diazoxide unresponsive HH requiring near-total pancreatectomy whilst milder variants cause diazoxide-responsive HH [25]. In some of the milder cases a bi-phasic phenotype has been reported whereby HH remits in childhood and diabetes is diagnosed in adulthood [26].
Glutamate dehydrogenase (GLUD1)
Gain of function mutations in GLUD1 gene, which encodes the mitochondrial enzyme glutamate dehydrogenase (GDH), cause leucine-sensitive HH (OMIM #606762) [27]. Within the pancreatic β-cell, leucine can activate GDH to catalyze the oxidative deamination of glutamate to α-ketoglutarate and ammonia. A-ketoglutarate then enters the tricarboxylic acid cycle (TCA) which generates ATP, ultimately leading to insulin exocytosis. Disease-causing variants in GLUD1 cluster around allosteric binding sites and act to reduce the sensitivity of GDH to inhibition by GTP and ATP [28]. This loss of regulation leads to an increase in the activity of GDH, a subsequent increase in the amount of α-ketoglutarate entering the TCA cycle and consequently an unregulated insulin secretory response.
Individuals with GLUD1 variants usually present with a milder form of HH that is often diagnosed outside of the neonatal period and is diazoxide-responsive [27]. In some patients dietary protein restriction may also be required. A consistent feature of this disorder is the presence of plasma ammonium concentrations raised two to three times the upper limit of normal. The presence of persistent hyperammonemia, in most but not all patients with GLUD1 variants [29], has led to this subtype of hyperinsulinism being referred to as Hyperinsulinism/Hyperammonemia (HI/HA) syndrome (OMIM #606762). An increased risk of epilepsy has also been observed in individuals with disease-causing GLUD1 variants [29].
In the majority of cases GLUD1 variants arise de novo, with no family history of hyperinsulinism. In keeping with their dominant nature, 50% of future offspring of affected individuals are at risk of inheriting the variant and developing HH.
Glucokinase (GCK)
Within the pancreatic β-cell glucokinase (GCK) plays a key role in linking insulin secretion to a glucose challenge by facilitating the phosphorylation of glucose to glucose-6-phosphate, the first step in glycolysis. Heterozygous gain-of-function variants in GCK cause HH by increasing the affinity of GCK for glucose which then acts to lower the threshold for glucose-stimulated insulin secretion [30] (OMIM #602485).
Individuals with gain-of-function GCK variants will often have a dominant family history of HH. The absence of a family history should however not preclude testing as de novo variants have been reported [31]. Variability in the severity of HH is also observed both in terms of age at presentation, which can range from birth to adulthood, and treatment response [31, 32]. Whilst the majority of individuals are successfully treated with diazoxide, some patients have medically unresponsive HH and require near-total pancreatectomy [32]. Although these differences in phenotype are likely to correlate with the functional severity of the variant, phenotypic variability within families with the same variant has been observed and is likely to be a consequence of genetic background and/or environmental factors [33].
Hydroxyacyl-coenzyme a dehydrogenase (HADH)
HADH encodes 3-Hydroxyacyl-coenzyme A dehydrogenase, an intramitochondrial enzyme that catalyzes the penultimate reaction in the β-oxidation pathway. Loss-of-function variants in HADH result in loss of interaction between HADH and glutamate dehydrogenase [34, 35]. This in turn leads to an increase in glutamate dehydrogenase activity, a subsequent rise in intracellular ATP and upregulated insulin secretion [36].
Patients with HADH disease-causing variants present with diazoxide-responsive protein-induced HH [3638](OMIM #609975). The severity in phenotype ranges from mild late onset hypoglycemia to severe neonatal hypoglycemia. In some patients there are raised plasma concentrations of 3-hydroxybutyrylcarnitine and urinary 3-hydroxyglutaric acid [36]. Loss- of- function variants in HADH are recessively inherited and this is the most common genetic subtype of HH in consanguineous individuals [39].
Hepatocyte nuclear factors (HNF4A and HNF1A)
The hepatocyte nuclear transcription factors, HNF1A and HNF4A, play crucial roles in glucose-stimulated insulin secretion as evidenced by the identification of loss-of- function variants in these genes in individuals with maturity-onset diabetes of the young (MODY)(OMIM #125850), an autosomal dominant form of diabetes typically diagnosed before the age of 25 years [40]. HNF4A variants were reported to cause a bi-phasic phenotype in individuals presenting with macrosomia and transient HH during the neonatal period and diabetes in later life [41]. The duration of HH varies markedly with some patients treated with intravenous glucose infusion for 1–2 days, yet others require diazoxide treatment for up to 11 years [4244]. HH with Fanconi syndrome, a renal tubular dysfunction, has been reported in at least 8 patients with HNF4A HH, all individuals have the p.R76W variant suggesting that this is a mutation specific phenotype [4548]. A few cases with HNF1A variants and transitory neonatal hypoglycemia have also been reported [45, 49].
Whilst a dominant family history of macrosomia, neonatal hypoglycemia and/or young onset diabetes can help to guide genetic testing for this condition, the absence of affected family members should not preclude analysis of HNF4A as de novo disease-causing variants have been reported [43].
Solute carrier family 16 member 1 (SLC16A1)
SLC16A1 gene encodes the monocarboxylate transporter (MCT1) which transports the insulin secretagogues pyruvate and lactate. Under normal physiological conditions SLC16A1 is not expressed in the β-cell thus preventing insulin from being secreted in response to lactate and pyruvate. Rare activating dominant variants result in the expression of MCT1 in the β-cell leading to pyruvate-stimulated insulin secretion following exercise [50, 51] (OMIM #610021). For patients with exercise-induced HH, treatment is not usually necessary as hypoglycemic episodes may be prevented by avoiding strenuous exercise [52].
Uncoupling protein 2 (UCP2)
UCP2 gene encodes an inner mitochondrial carrier protein UCP2 which is widely expressed in tissues, including pancreatic islets [53, 54]. UCP2 can inhibit ATP generation by causing proton leak across the inner mitochondrial membrane and negatively regulates glucose-mediated insulin secretion [54]. Inactivating heterozygous mutations in the UCP2 gene can enhance glucose oxidation and increase intracellular ATP synthesis leading to HH [54]. UCP2 mutations can present with either transient or prolonged HH [55]. However, in a recent published study, no mutations were detected in the UCP2 gene among 206 diazoxide responsive patients [56], suggesting that the role of UCP2 in HH needs further investigation.
Hexokinase 1 (HK1)
HK1 encodes the hexokinase HK1, which catalyzes the phosphorylation of glucose to glucose-6-phosphate as substrate for glycolysis. A family with dominant gain-of-function mutation in the HK1 gene has been reported with “idiopathic hypoglycemia of infancy” [57]. In vitro studies evaluating pancreatic β-cells from CHH patients have shown inappropriate expression of HK1. These pancreatic tissues showed functional KATP channels with inappropriate secretion of insulin at low plasma glucose concentrations (1 mmol/L) [58].
Phosphoglucomutase 1 (PGM1)
PGM1 is involved in glycogen metabolism and is responsible for reversible conversion of glucose-6-phosphate to glucose-1-phosphate. PGM1 gene encodes the enzyme PGM1 and recessive loss-of-function mutations in PGM1 cause hypoglycemia [59].
Children with PGM1 mutation presented with postprandial HH and fasting hyperketotic hypoglycemia [59].
Phosphomannomutase 2 (PMM2)
Recessively inherited variants in PMM2, which encodes a key enzyme in N-glycosylation, have been identified in individuals with HH and polycystic kidney disease. In all individuals a c.-167G > T promotor variant was identified that was either homozygous or in trans with a coding variant [60].
The majority of patients with PMM2 variants present with macrosomia at birth and hypoglycemia in the first year of life. For many, hypoglycemia was the presenting feature and often manifested with seizures. Patients are responsive to treatment with diazoxide [60].
Forkhead box protein A2 (FOXA2)
Mutations in FOXA2 have been reported to cause hypopituitarism, CHH and endoderm-derived organ abnormalities. These children have a unique clinical phenotype of hypopituitarism, CHH, dysmorphic features, and liver, pancreas, heart and gastrointestinal abnormalities [61, 62].
Calcium voltage-gated channel subunit alpha1 D (CACNA1D)
CACNA1D gene encodes an L-type voltage-gated calcium channel which is expressed in pancreatic β-cells and regulates insulin secretion. Mutations in CACNA1D have been reported to cause HH, heart defects and severe hypotonia [63].
Eukaryotic translation initiation factor 2 subunit 3 (EIF2S3)
Three cases published with variant in EIF2S3 present an unusual dysregulation of glucose fluctuating between diazoxide-responsive HH and postprandial hyperglycemia, along with learning difficulties and hypopituitarism [64].

4.3 Other forms

4.3.1 Postprandial forms of HH

In postprandial hyperinsulinemic hypoglycemia (PPHH), hypoglycemia is induced hours after meal intake due to inappropriate/exaggerated insulin secretion in response to the meal. The most common cause is “dumping” syndrome in infants/children who have undergone Nissen fundoplication/gastric bypass [65, 66]. Children with PPHH after Nissen fundoplication have an abnormally exaggerated secretion of the insulin secretagogue glucagon-like peptide 1 (GLP-1) which may contribute to the exaggerated insulin surge and resultant hypoglycemia [67].
PPHH is also reported in the insulin autoimmune syndrome leading to development of insulin binding autoantibodies in children who were not previously exposed to exogenous insulin [68].

4.3.2 Other causes of HH

Insulinoma is a rare cause of hyperinsulinism and must be considered in older children or adolescents presenting with HH [69]. A detailed family history of tumors especially like insulinoma, is relevant as it can be part of multiple endocrine neoplasia syndrome type 1 (MEN1).
Munchausen by proxy leading to exogenous administration of insulin or anti-diabetic drugs such as sulphonylureas can present as factitious HH. This has led to misdiagnosis and consequent pancreatectomy [70].

4.3.3 Syndromes associated with HH

A large number of syndromes are associated with HH, with Beckwith-Wiedemann syndrome (BWS) being one of the commonest [71]. Table 2 lists the syndromic associations with HH, although the exact mechanism for HH is still not well understood in all the conditions. Most syndromic forms are diazoxide-reponsive and HH resolves overtime.
Table 2
Syndromic forms of HH - Various developmental syndromes have been described with the gene/s linked to the condition and the common clinical features [203]
SYNDROME NAME
GENETIC ETIOLOGY
gene (location)
CLINICAL CHARACTERISTICS
Pre- and postnatal overgrowth (Macrosomia)
  Beckwith-Wiedemann
(11p15)
Macroglossia, abdominal wall defects, ear lobe pits/ creases, hemihypertrophy, tumor risk
  Sotos
NSD1 (5q35)
Macrocephaly, frontal bossing, pointed chin, developmental delay, tumor risk
  Simpson-Golabi-Behmel
GPC3 (Xq26), GPC4 (Xp22)
Coarse facial features, broad feet, polydactyly, cryptorchidism, hepatomegaly, tumor risk
  Perlman
DIS3L2 (2q37)
Inverted V-shaped upper lip, prominent forehead, developmental delay, hypotonia, tumor risk
Postnatal growth failure (short stature)
  Kabuki
KMT2D (12q13), KDM6A (Xp11.3)
Arched eyebrows, long eyelashes, developmental delay, fetal finger pads, scoliosis, heart defects, hypotonia
  Costello
HRAS (11p15)
Deep palmar/plantar creases, developmental delay coarse facial features, heart abnormalities, papillomas, tumor risk
Chromosomal abnormality
  Mosaic Turner
(Loss of X in some cells)
Milder Turner syndrome phenotype (short stature, coarctation of aorta, gonadal dysgenesis)
  Patau
Trisomy 13
Developmental delay, microphthalmia, heart & neural defects
Congenital disorders of glycosylation
  Types 1a, 1b, and 1d
PMM2 (16p13.​2), MPI (15q24.​1), ALG3 (3q27.​1)
Developmental delay, hypotonia, growth failure
Contiguous gene deletion affecting the ABCC8 gene
  Usher
11 genes
Hearing loss, visual impairment
Abnormalities in calcium homoeostasis
  Timothy
CACNA1C (12p13.​33)
Long QT syndrome, syndactyly, developmental delay, immune deficiency
Insulin receptor mutation:
  Insulin resistance syndrome (leprechaunism)
INS (19p13)
Hypo- and hyperglycemia, pre- and postnatal growth restriction, elfin-like features, hirsutism
Other Syndromes:
  Congenital central hypoventilation syndrome
PHOX2B (4p13)
Central hypoventilation, “box-shaped” face, neurocristopathies (Hirschsprung disease, tumor risk)

5 Pathophysiology of HH - histological subtypes

There are three histological forms of CHH (Fig. 2); Focal form (F-CHH), Diffuse form (D-CHH) and atypical. The clinical presentation appears to be similar, although their molecular mechanisms are quite different. In most cases D-CHH is inherited in an autosomal recessive manner whereas the F-CHH is sporadic [22, 72].

5.1 Focal form of CHH (F-CHH)

Focal lesions occur when the abnormal pancreatic β-cells are localized to a single specific location in the pancreas. They are the result of two unique events, first the inheritance of a paternally inherited ABCC8 or KCNJ11 mutation at 11p15.1, and secondly the loss of the corresponding maternal allele within the focal lesion. This causes an imbalance in the expression of imprinted genes such as the maternally expressed tumor suppressor gene H19 and CDKN1C, and the paternally expressed growth factor IGF-2, at 11p15.5 [73] leading to β-cell hyperplasia. Within the focal islet cell lesion there is adenomatous hyperplasia and abnormally large nuclei in the affected cells.
A duplication of the paternal allele located on chromosome 11 has also been found in some patients with F-CHH [74]. Outside the lesion, normal endocrine tissue with small nuclei exists. These cells appear to have less cytoplasm and diminished proinsulin production [7578].
Patients who have a heterozygous paternally inherited mutation in ABCC8 or KCNJ11 could have F-CHH, which accounts for 30–40% of all CHH cases [79]. F-CHH is confirmed by a fluorine-18 dihydroxyphenylalanine-positron emission tomography (18F-DOPA-PET) scan, which can show the presence of a focal lesion and determine its location with diagnostic accuracy [80].

5.2 Diffuse form of CHH (D-CHH)

The D-CHH form occurs when all the islet cells in the pancreas are abnormal [77, 81]. Patients with a homozygous recessive or a compound heterozygote mutation in ABCC8 or KCNJ11 present with D-CHH. These patients are usually medically unresponsive, and this histological form accounts for 60–70% of all CHH cases. Most islets throughout the entire pancreas are affected with the presence of large hyperchromatic nuclei [7578, 81].

5.3 Atypical forms of HH

Histologically atypical forms of CHH are categorized when the pancreatic morphology does not fit into the F-CHH or D-CHH types and are a mosaic pattern of the two [73, 82]. The islets can either be enlarged or shrunken. Some cases have been cured with a lesionectomy whilst others also require medical management. However, to date only one patient has been described with a ABCC8 nonsense mutation (Q54X) causing this histological form of CHH [82]. Studies have shown that the heterogeneous expression of β-cells HK1 in the atypical CHH may be causing the abnormal insulin release [83, 84].

6 Clinical presentation of HH

HH usually manifests in infancy or early childhood, although some patients can present during adolescence or adulthood. Signs and symptoms of hypoglycemia are non-specific during the neonatal period (poor feeding, jitteriness, irritability), challenging its diagnosis. At later ages, symptoms can be easier to recognize and can be classified as adrenergic (hunger, pallor, tachycardia, tremor, diaphoresis) and neuroglycopenic (tiredness, blurred vision, confusion, coma and even death).
Risk factors for HH need to be looked for in the perinatal history. These include maternal intrapartum administration of hypoglycemic agents, stressful delivery, large/low birth weight, and neonatal polycythemia or jaundice, among others. Examination of the baby will search for macrosomia/leanness, careful phenotypic characterization to identify syndromes, features of cardiomyopathy (due to glycogen storage in HH), hepatomegaly present in metabolic conditions (also present in HH), and midline abnormalities (including genitalia) to exclude hypothalamic-pituitary deficits. Data in the family history could suggest a genetic inheritance, therefore anamnesis must enquire for consanguinity, cases of diabetes mellitus, hypoglycemia, seizures and unexplained deaths [85].

7 Diagnostic investigations of HH

Any child requiring intravenous glucose load greater than 8 mg/kg/min (normal requirement is 4-6 mg/kg/min) to avoid hypoglycemia, can essentially be labeled as HH [86]. The timing of hypoglycemic events with regard to meals will give insight into the intrinsic mechanism. HH typically manifests during brief periods of fasting, however certain types can be manifest soon after meal ingestion (protein induced HH) or hours after meal ingestion (postprandial HH). Should there be a clear link between physical activity and occurrence of hypoglycemia, then exercise induced HH should be excluded.

7.1 Biochemistry

Biochemical interpretation will only be possible if the critical sample is taken adequately at the time of hypoglycemia (≤3.0 mmol/l), ideally in a controlled fasting setting. HH will demonstrate detectable C-peptide or insulin in the face of hypoglycemia, with simultaneously low/undetectable alternative substrates (ketones and fatty acids) [86, 87]. Of note, the concentration of insulin does not parallel the degree of severity of the condition [88, 89]. Table 3 defines the diagnostic criteria for HH. If a particular mechanism is suspected to trigger HH (exercise, protein, carbohydrate etc) specific stimulation tests will be carried out:
Table 3
Diagnostic criteria for HH - The cut-off values for each analyte to aid in the diagnosis of HH. IGFBP-1: Insulin growth factor binding protein-1. HI/HA: Hyperinsulinemic hypoglycemia hyperammonemia syndrome. HADH: short chain L-3-hydroxyacil-CoA dehydrogenase. Im: intramuscular. Iv: intravenous. Sc: subcutaneous *[90, 200, 203]¥
Serum analyte
Result in patients with HH
Blood glucose < 3.0 mmol/l (54 mg/dl) and:
  Insulin
Detectable
  C-peptide
Detectable (≥0.5 ng/mL¥)
  Free fatty acids
Low or suppressed (<1.5 mmol/l* or < 1.7 mmol/l¥)
  Ketone bodies
Low or suppressed (3-β-hydroxybutyrate <2 mmol/l* or < 1.8 mmol/l¥)
  IGFBP-1
Low (≤110 ng/mL¥) as insulin negatively regulates IGFBP-1 expression
  Ammonia
Normal. Can be raised in HI/HA syndrome
  Hydroxybutyrylcarnitine
Normal. Raised in HH due to HADH mutation
  Cortisol, Growth hormone
Raised. Generally Cortisol >20 μg/dL [500 nmol/L]; growth hormone >7 ng/mL - younger children might have poor counter-regulatory response
  Amino acids and urine organic acids
Normal. Leucine, isoleucine and valine may be suppressed in HH
  Proinsulin
>20 pmol/l
Additional information when diagnosis of HH uncertain:
  Glucose infusion rate
>8 mg/kg/min to achieve euglycemia
  Im or iv glucagon administration or
sc octreotide administration
>1.5 mmol/L or 27 mg/dl (Positive glycemic response)

7.1.1 Protein load test

In this test leucine or a combination of aminoacids is administered enterally. Evidence of raised insulin concentrations at the time of hypoglycemia will confirm the clinical suspicion of protein sensitive HH [29].

7.1.2 Oral glucose tolerance test / mixed meal test

Enteral administration of a mixed meal or oral glucose load followed by hypoglycemia with a detectable insulin within 2–5 h, indicates incretin mediation leading to postprandial HH. In adults the prefered test is the standardized hyperglucidic breakfast [91], which has not yet been standardized in children.

7.1.3 Exercise test and pyruvate load

Biochemical evidence of HH during these tests indicate a lactate/pyruvate mechanism triggering exercise induced HH [92].

7.1.4 Fructose load test

Individuals with ketotic hypoglycemia after fructose ingestion should undertake a fructose load test to identify hereditary fructose intolerance and fructose-1,6-diphosphatase deficiency, and this can be followed by genetic testing.

7.1.5 The role of real-time continuous glucose monitoring (CGM)

CGM measures interstitial glucose concentrations indicating the direction, magnitude, frequency, duration and causes of fluctuations in these. There is now a study on its accuracy in children with HH [93], as this can significantly improve the quality of life of these patients.

7.2 Imaging

7.2.1 Recent advances in diagnostic imaging of HH

As is not possible to differentiate between F-CHH and D-CHH based on clinical presentation and biochemical features, pre-operative differentiation of both subtypes is critically important. Conventional radiological imaging such as Magnetic Resonance Imaging (MRI) and Computerized Tomography (CT) scans fail to localize the focal lesions. Genetic analysis for mutations in ABCC8/KCNJ11, with combination of recently described Fluorine 18 L-3, 4- dihydroxyphenyalanine positron emission tomography (18F-DOPA-PET-CT) scanning, allows differentiation between F-CHH and D-CHH with a high sensitivity (88%) and specificity (94%) [9498], with an accuracy of 100% [99102]. The principle of 18F-DOPA-PET analysis is based on the selective uptake of L-DOPA by β-cells and its conversion into dopamine by DOPA decarboxylase enzyme, which is expressed in the pancreatic β-cells. Thus, imaging with 18F-DOPA-PET-CT should be performed in all patients who are thought to have F-CHH. Some patients have been described with atypical histological forms, which are either atypical focal, atypical diffuse or ectopic β-cell hyperplasia and do not show classical features of F-CHH or D-CHH [103]. Demonstration of increased activity of DOPA decarboxylase by 18F-DOPA-PET in combination with an enhanced CT imaging can successfully differentiate diffuse and focal β-cell hyperplasia. Therefore, this technique has radically changed the surgical approach to patients with medically unresponsive HH [102, 104].
A dosimetry study of 18F-DOPA derived from the PET-CT images in ten infants (median age 4.84 weeks) with HH suggested that a modest radiation dose (0.30 ± 0.04 mSv/MBq) was adequate [105]. However, there have been recent reports of its inaccuracy in precisely detecting focal lesions [106, 107]. Glucagon-like-peptide-1 (GLP-1) receptor analogs are the latest agents being used in the detection of insulinomas in adults [108, 109]. Similarly, this isotope is being investigated in children to detect focal lesions.

7.2.2 Imaging for insulinoma

Most insulinomas are benign (islet-cell tumors) and usually very small (<2 cm) making it difficult to localize with the current imaging techniques. Various non-invasive techniques have been used for the detection of insulinomas e.g., transabdominal ultrasonography, spiral CT, MRI, 111In-pentetreotide imaging, and 18F-l-dihydroxyphenylalanine PET. In difficult cases, invasive procedures like endoscopic ultrasonography or selective arterial calcium stimulation test with hepatic venous sampling have also been used [110].
A systematic review and meta-analysis of (68)Ga-DOTATATE compared with octreotide and conventional imaging documented a high sensitivity (90.9%) and specificity (90.6%) of (68)Ga-DOTATATE in detecting neuroendocrine tumors [111]. Various Gallium based isotopes have also been used to detect insulinoma in adults [108, 112].

8 Management of HH

Maintaining normoglycemia (blood glucose >3.5 mmol/l) is paramount to avoid hypoglycemic brain injury in view of the hypoketotic nature of HH [113]. This may be achieved by increasing glucose administration (feeds or intravenous (iv) fluids), stimulating endogenous glucose release (glucagon administration) or suppressing insulin release from the β-cell (diazoxide, octreotide, nifedipine). Oral feeds with additional glucose polymer in combination with iv fluids can also be used to maintain normoglycemia. The traditional and new drugs available for the management of HH are summarized in Table 4.
Table 4
Standard and novel drugs used in the management of HH - Medications used for the treatment of HH, along with their dose, mechanism of action and side effects [114116, 134, 140, 201]
Medication
(route of administration)
Total daily dose
Action mechanism
Side effects
Standard drugs
Diazoxide (enteral)
5-20 mg/kg/day (divided in 3 doses)
Binds to the SUR1 subunit of intact KATP channels, opening the channel and inhibiting insulin release
Common: Fluid and sodium retention, hypertrichosis, anorexia.
Rare: Cardiac failure, pulmonary hypertension**, blood dyscrasia, hyperuricemia, paradoxical hypoglycemia
Chlorothiazide (enteral)
7-10 mg/kg/day (divided in 2 doses)
Synergy with diazoxide over KATP channels inhibiting insulin secretion. Prevents fluid overload
Hyponatremia, hypokalemia
Glucagon (sc/im bolus; sc/iv infusion)
Bolus: 0.02 mg/kg/dose
Infusion: 2.5–10 mcg/kg/h
Stimulates glycogenolysis, gluconeogenesis, ketogenesis, lipolysis
Skin rash, vomiting. Paradoxical rebound hipoglycemia if dose >20mcg/kg/h (high dose stimulates insulin release)
Octreotide
(sc)
5–40 mcg/kg/day (divided in 3–4 doses or continuous infusion)
Activation of SSTR-2 and SSTR-5. Stabilisation of KATP channel, reduces calcium entry in β-cell, inhibition of insulin secretion. Inhibitition of INS promoter.
Acute: Abdominal discomfort, vomiting, diarrhea, anorexia, hepatitis, transaminasemia, long QT syndrome, necrotizing enterocolitis, tachyphylaxis.
Long-term: Cholelithiasis, intestinal hypomobility, suppression of GH and TSH
Nifedipine (enteral)
0.25–2.5 mg/kg/day (divided in 2–3 doses)
Blockage of β-cell calcium channel activity, leading to inhibition of insulin exocytosis
Hypotension
Acarbose (enteral)
6.25–300 mg/day (divided in 3 doses – before main meals)
Inhibits intestinal α-glucosidase (cleaves polysaccharides to monosaccharides)
Intestinal discomfort, diarrhoea, flatulence, raised transaminases
Novel drugs
Lanreotide and long-acting octreotide
(deep sc or im)
30–60 (max 120*) mg/dose
(every 4 weeks)
Like octreotide. High affinity for SSTR 2 & 5, and reduced affinity for SSTR 1, 3 & 4
Same as octreotide. Pain at injection site. No long-term data available yet.
mTOR inhibitors
(enteral)
Starting dose: 1 mg/m2/day (divided in 2 doses). Adjust dose aiming for blood concentrations 5-15 ng/ml
Inhibits mTOR complex 1. Inhibits β-cell proliferation and insulin secretion. Posible induction of peripheral insulin resistance
Immune suppression, hyperlipidemia, hypertransaminasemia, mucositis, thrombocytosis
Sc: subcutaneous. Im: intramuscular. Iv: intravenous. SSTR: Somatostatin receptor. INS: insulin gene. SUR1 = sulfonylurea receptor 1. KATP = ATP-sensitive potassium channel.
**[115, 116]
*90-120 mg every 4 weeks [134]. The starting dose of Lanreotide autogel 30 mg has been found to be effective [132, 133]

9 Current medical management

9.1 Acute management of hypoglycemia

If the oral route is unavailable or glycemia is not improving despite oral glucose (glucose gel/glucose containing drinks or tablets), then an iv dextrose bolus (2 ml/kg 10%Glucose) needs to be administered followed by continuous iv glucose infusion (>8 mg/kg/min). If there is persistence of hypoglycemia, hypoglycemic seizure or inadequate iv access, then an intramuscular or subcutaneous bolus (1 mg) or infusion of glucagon can be life saving as it causes immediate release of glycogen stores from the liver leading to a temporary improvement in blood glucose concentrations. [117].
As high iv glucose concentration is usually needed for weeks, insertion of a central venous access supports in managing glycemia safely.

9.2 Long-term pharmacological agents

Medical therapies can be tried to wean the infant off iv support and achieve a near-normal feeding pattern. Surgical therapy is considered when trial of medical therapies is unsuccessful or in case of established F-CHH.
HH drug responsiveness is defined by: 1) normal feeding frequency and volume, 2) fasting capability adequate for age whilst maintaining euglycemia, 3) low/suppressed serum insulin level at the end of the fast, and 4) appropriate generation of ketone bodies and fatty acids by the end of the fast [117].
Diazoxide
The first drug of choice is diazoxide [86], which requires a functional KATP channel to bind onto and is hence ineffective in D-CHH due to an inactivating KATP mutation and in most cases of F-CHH. Most other forms of HH and protracted HH secondary to risk factors such as IUGR and perinatal asphyxia respond to treatment with diazoxide. Due to its fluid retaining properties, diazoxide therapy must be used with caution in infants with HH who are often receiving large volumes of iv/oral fluids to maintain normoglycemia. Fluid restriction prior to starting diazoxide is commonly practiced, along with concomitant use of a thiazide diuretic such as chlorothiazide, which also has a synergistic action over the KATP channels.

9.2.1 Medical management of diazoxide unresponsive D-CHH

Nifedipine
Authors suggest triyng it before heading to pancreatectomy [118] as there have been isolated case reports on successful treatment of HH with nifedipine [119121] but experience of large HH centers has generally been disappointing [117, 122, 123]. It is sometimes considered as an add-on drug in partial diazoxide/octreotide resistance, and/or following partial pancreatectomy [122, 124] but rarely used as monotherapy.
Octreotide
Octreotide is a second-line treatment for diazoxide-insensitive patients [125, 126]. It may be used in combination with diazoxide and glucagon in cases with partial diazoxide response, or often combined with frequent feeding which may require a gastrostomy to enable high calorie bolus feeding during the day and overnight continuous feeds. As octreotide binds to the somatostatin receptors SSTR-2 and SSTR-5, prolonged use may develop into drug desensitizing caused by internalisation of the receptors [122].
Glucagon
Iv or subcutaneous (sc) glucagon [127] may be helpful during the initial stabilization period and before surgery. However, this treatment has not been of long-term benefit. It can also be administered (alone or in combination with Octreotide) to stabilize blood glucose concentrations in the acute management and prevent near-total pancreatectomy in infants with HH [128]. Continuous sc glucagon infusion can frequently complicate by catheter obstructions occurring daily or 2–3 times per week [128].
Acarbose
For postprandial HH the first approach is diet modification. This includes: frequent feeds of long-acting carbohydrates, abundant protein and supplements of fibre and fat emulsions [129]. If hypoglycemia persists, acarbose is the preferred medical option as it slows the absorption of glucose into the blood stream, thus avoiding a glycemic peak followed by insulin release [130].

10 Novel medical therapies

10.1 Long-acting somatostatin analogues

Lanreotide and Long-acting release octreotide (LAR-Octreotide) are two formulations used in few numbers of patients with HH [131136] reported not only to be useful in the management of HH in children, but potentially displaying a more stable glycemic control than octreotide [134]. Lanreotide has also been proven useful in managing inoperable F-CHH [137].

10.2 mTOR inhibitors

The intracellular mTOR pathway is involved in β-cell growth and altered insulin secretion in patients with insulinoma [138]. Therefore, mTOR inhibitors such as sirolimus (formerly known as rapamycin) and everolimus have been used to treat this tumor. Although the exact mechanism of sirolimus in HH still needs to be elucidated, it has been hypothesized that the mTOR complex 1 may be overactivated in D-CHH [139]. The first study reported the use of sirolimus in 4 children with severe D-CHH who achieved glycemic control avoiding pancreatectomy and with no major side effects [140]. This has been followed by various case reports of HH in children of various ages who have benefitted from sirolimus [141145]. However, there have been reports about its severe side effects as well as poor response in others [146149]. Our center has tried sirolimus on 22 patients with various forms of HH, out of which 21 showed glycemic response, however 19 patients developed side effects, recurrent and frequent infections being the most common infections [150]. There is no clear correlation between genetic etiology and response to sirolimus.

11 Potential novel therapies – The future

11.1 GLP-1 antagonists

GLP-1 acts on the β-cell promoting its proliferation and stimulating insulin release [151]. Mouse models with KATP channel defects leading to HH, significantly improved their glucose concentrations when treated with GLP-1 receptor antagonist (exendin-9-39) [152]. Infusion of exendin-9-39 has been tried in 9 adult patients with HH due to KATP mutations [153], where all patients demonstrated increased fasting mean glucose and glucose area under the curve, raising expectation for its near-future use in children with this condition.

11.2 Pharmacological trafficking chaperones

Some mutations in the ABCC8 gene prevent trafficking of parts of the SUR1 channel from the endoplasmic reticulum to the surface of the cell. Carbamazepine [154] and sulfonylureas [155] – glibenclamide and tolbutamide - are KATP channel inhibitors and chaperones that have proven to amend channel trafficking defects in many ABCC8 mutations by allowing intersubunit interactions between SUR1 and Kir6.2 [156]. This has been now confirmed in mouse functional studies as well [157] reinforcing its potential for clinical use.

11.3 Glucagon – New formulations and delivery

Subcutaneous glucagon infusion via pump has been used as long-term treatment for HH patients at home [158]. Practical issues with its administration remain unsolved as it can cause mechanical obstruction due to its formulation. However, a stable form of glucagon formulation could be a potential useful tool that is being explored [159, 160].

12 Surgical management

12.1 Surgery for F-CHH

Removal of the affected part of the pancreas achieving complete cure is the surgical aim for the F-CHH. A multidisciplinary team (endocrinology, radiology, and histopathology) should be involved in managing patients with F-CHH and D-CHH [161, 162]. In most cases, pre-operative 18F-DOPA PET-CT helps exact localization of the focal lesion and aids the surgeon. Intra-operative biopsies are important to ensure complete excision with histological confirmation of clear margins. Laparoscopic lesionectomy is the preferred surgical approach when the focal lesion is easily accessible (i.e. body or tail of pancreas) offering the benefit of shorter post-operative care [163]. Focal lesions that are difficult to access such as in the head of the pancreas usually require open laparotomy for resection of most of the pancreatic head and Roux-en-Y pancreaticojejunostomy [164].

12.2 Surgery for D-CHH

Near-total pancreatectomy (95–98% of the pancreas), the only option for medically unresponsive D-CHH has largely been reported to have unsatisfactory outcomes [165, 166]. In a large study, nearly 59% of patients that underwent near total pancreatectomy continued to experience hypoglycemia, though it was usually easier to manage with dietetic/medical therapy. Post-operatively, hyperglycemia was common as well with increasing incidence as age progressed and a 100% incidence by 13 years of age [166]. Complex glucose derangements with persisting fasting hypoglycemia and post-prandial hyperglycemia were also reported in 35% of patients. Exocrine pancreatic insufficiency is also common following near-total pancreatectomy for D-CHH [166]. Laparoscopic surgery is now the preferred option as opposed to the traditional open approach [161, 167].

12.3 Surgery for insulinoma

Surgical removal of the tumor is the mainstay therapy for childhood and adult insulinoma, and its overall cure rate is up to 98% [168, 169]. Prognosis depends on the tumor stage at the time of presentation and success rate of complete resection. The mode of surgery depends on the tumor size, localization, and metastatic characteristics [69, 170]. For small benign tumors with no metastasis that are located at least 2–3 mm from the main pancreatic duct, a limited enucleation should be performed [171]. A tumor that is invading the pancreatic duct or great vessels with risk of malignancy and lymph node invasion, and that is compressing the distal pancreatic duct, might require a more extensive surgical resection. The surgical resection procedure depends on the site of the insulinoma and includes mid-body pancreatectomy, distal pancreatectomy, or pylorus-preserving Whipple procedure [171, 172].

13 HH monitoring and follow up

Children with HH need regular monitoring of their blood glucose at home. The follow-up of these children requires involvement of a multidisciplinary team including pediatric endocrinologist, dietitian, nurse specialist, clinical psychologist, developmental pediatrician and pediatric surgeon. Severity and treatment requirement should be assessed periodically in all children on medical therapy and the dose of medications should be optimized based on their glucose monitoring and fast tolerance test. It has been reported that for most patients with KATP channel mutations who are managed by medical treatment only, severity is reduced over time [173]. Children need to be monitored for the side effects of the HH medications [135, 146, 147, 174, 175]. Also, the majority of children with severe HH are on high carbohydrate feeds administered via gastrostomy by bolus and/or continuous feeds, hence the importance in receiving support from the community i.e. nursery, school, home etc. in these group of children. Post-surgical patients require periodical monitoring including stool elastase, capillary glucose measurements and oral glucose tolerance test looking for complications (hypoglycemia, diabetes and exocrine pancreatic insufficiency) [165, 166]. Various studies evaluating the long-term outcome of patients with HH have reported a high frequency of neurodevelopment delay and various neurological disorders, including epilepsy and microcephaly [5, 176, 177]. This group of children may need a special education plan devised with the help of the educational psychologist and developmental pediatrician.

14 Transition into adult service

Young adults with HH have complex management requirements that are best supported using a multidisciplinary approach [178]. The move from pediatric to adult services may be a challenging time for adolescents transitioning to self-management [179].
A transition service aims to bridge this changeover in care, and support the adolescent [180] in terms of their health needs, but also psychosocial development, including ability to achieve independence and establish adult relationships [181, 182]. This specific age group are recognized to have barriers that prevent optimal self-management, including [183];
  • heightened concerns about peer relationships and social interactions,
  • frustration and fatigue from the management of a chronic illness,
  • incomplete knowledge and understanding of chronic disease management,
  • inclination towards risk-taking and
  • difficulties in the transition to self-management.
People with chronic conditions experience differences in the clinical management approach of pediatric and adult care settings during the transition process [182]. Pediatric teams may overlook the growing independence of the individual, however conversely, the encouragement to take responsibility from adult care providers may lead to physical, psychological and social development being neglected [182, 184, 185].
Consequently, young adults often feel misplaced in adult services and this has been shown to lead to lower rates of follow-up appointments, attendance and medication compliance [181, 186].
Discharge to a non-specialist adult team carries with it some challenges, including lack of awareness about this rare condition and its complex on-going health-needs. In addition, non-specialist teams may not be best placed to undertake discussions around pre-conception counselling in those with a confirmed genetic etiology.

14.1 Aims of the transition service

Recognizing the core attributes of this age group, a transition service for patients with HH may be established, with the following aims and in line with established NICE guidance [178].
1.
Supporting education: Establish and support understanding of HH. Where genetic causes are identified, patients should be aware of this and have an understanding of its inheritance.
 
2.
Empowering self-management: To support handover of care to the affected individual from the carers [187], by encouraging shared decision-making and empowering self-management, with an ultimate aim of self-management [188].
 
3.
Addressing parental and care-giver concern [189]: Establishing an understanding between the young person and their carer of how aspects of self-care might be jointly undertaken and transferred is an important component of managing distress, expectations and, ultimately, transition [190].
 
4.
Addressing on-going medical issues: Table 5 summarizes some of the medical issues that may be addressed in a transition service. The risk and progression to diabetes is variable and appropriate treatments for diabetes may depend on both the mutation causing HH and the extent of β-cell failure.
 
5.
Dealing with impaired awareness of hypoglycemia [191]: If found in severe cases of HH it can be formally assessed using the validated Clarke [192] or Gold score [193]. The evidence base for managing impaired awareness of hypoglycemia is limited in the context of HH [194] and so, care is needed in managing these individuals. CGM could be of use in some cases [191].
 
6.
Providing advice around driving: Supporting individuals at high risk of hypoglycemia or impaired awareness of hypoglycemia to apply for a driving license may be explored, and fully explaining driving regulations in relation to hypoglycemia is important for those starting to learn to drive [195].
 
7.
Providing support to higher educational institutions and those in employment [196, 197]: Flexibility may be required to allow time for testing, or additional breaks for calorie consumption.
 
8.
Pre-conception genetic counseling, where appropriate: In those with confirmed dominant mutations, ensuring adequate counseling around offspring, and accessing genetic counselors when appropriate, is important. Insulin modulates Sertoli cell function and young hyperinsulinemic patients have been found to have lower anti-Müllerian hormone and inhibin B secretion [198]. This could therefore influence testicular function and have a future impact on fertility, which still needs to be studied.
 
9.
Continued dietetic advice: On-going advice about dietary management of hypoglycemia, especially where there is protein-induced hypoglycemia, and replacement of calories with other macronutrients may be provided.
 
10.
Alcohol and recreational drug advice [199]: alcohol consumption can potentiate the risk of hypoglycemia and also impair awareness of hypoglycemia. Recreational drugs may also mask the adrenergic symptoms of hypoglycemia.
 
Table 5
Summary of medical issues encountered in adolescent and young adult patients with congenital hyperinsulinism and the intervention required.
Medical Issue
Support / Intervention needed
Confirmed mutation causing HH
  Symptom control
• Exploring precipitants
• Dietary interventions and advice
• Need for medical treatment e.g. diazoxide, calcium channel blockers, somatostatin analogues.
• Accessing appropriate technology e.g. real-time CGM, where appropriate
  Risk of diabetes
• Aware of diabetes symptoms
• Annual glucose checks
• Understands the risk of diabetes
  Managing diabetes in non-pancreatectomized individuals
• Establishing type of diabetes
• Impact of underlying genetic mutation
• Measuring endogenous insulin production to determine if insulin needed.
  Managing diabetes in pancreatectomized individuals
• Diagnosing and treating insulin-deficient diabetes in these individuals early on
• Ensuring life-long insulin and clearly aware of diagnosis
• Managing concurrent exocrine failure
• Loss of glucagon may also contribute to problematic hypoglycemia
• Ensuring access to appropriate diabetes technologies e.g. insulin pumps and continuous glucose monitoring
  Impaired hypoglycemia awareness
• Checking individuals know the symptoms of hypoglycemia
• Assessing awareness of hypoglycemia using validated scores e.g. Clarke or GOLD score.
• Considering adjunctive use of monitoring technologies such as real-time continuous glucose monitoring in those with hypoglycemia unawareness.
No mutation identified (in addition to above)
  Exploring a genetic diagnosis
• Ensuring panel of all genes tested
• Undertaking whole exome or whole genome sequencing studies to identify novel genes
• Re-characterizing type of hyperinsulinism and considering alternative diagnosis
  Counseling around diagnostic uncertainty
• Ensure adequately knowledgeable about their condition
• Symptom control
• Need to continue medical therapy
• Pregnancy

14.2 Developing the HH transition pathways

In line with NICE transition service guidelines [178], the optimal pathway for transitioning individuals with HH must be developed, summarized in Fig. 3. This includes:
  • Planning a service: Young people should be involved in designing the service.
  • Planning for transition, which should be appropriately timed for the individual.
  • Before Transition: young adults should have joint clinics with the pediatric and adult teams.

15 Concluding remarks and future directions

HH is a challenging condition to treat due to its heterogeneity. Despite diagnostic and therapeutic advances, HH remains an important cause of morbidity in children, still accounting for 26–44% of permanent intellectual disabilities, especially in neonatal-onset patients. The increasing use of NGS target panels, combined with clinical, biochemical and imaging findings allows differentiating the diagnostic management of children with F-CHH, surgically curable, from those with D-CHH, more conservatively treated with pharmacological and nutritional interventions [200].
There is now more research of use of CGM in children with HH which might help to detect early hypoglycemia leading to prompt management [93]. There have been reports of pitfalls of 18F-DOPA PET-CT in accurately detecting focal lesions in CHH [106, 173]. Glucagon-like-peptide-1 (GLP-1) receptor analogs are the latest agents being used in the detection of insulinomas in adults [108, 109] and are similarly being trialed in children to detect F-CHH.
Over the last few years, numerous medications have been tried in children with HH [201]. For instance long-acting somatostatin analogues and mTOR inhibitors (sirolimus) have been used in various groups of children with varied response [133136, 140142, 148]. There is potential use of insulin receptor antagonists as a therapeutic approach to control hypoglycemia in CHH [202, 203]. Also, reports have documented the use of a stable form of glucagon in adults with hypoglycemia [159, 160], however more clinical trials are required to prove its efficacy in children with HH.
Besides the development of new diagnostic tools and therapeutic agents, clinicians need to become involved in creating/potentiating HH transition clinics to provide optimal ongoing care into adulthood for these patients.

Compliance with ethical standards

Conflict of interest

Nick Oliver has received honoraria for advisory board participation or speaking from Dexcom, Roche diabetes, and Medtronic diabetes; support for education from Novo Nordisk and Takeda; and research funding from Dexcom and Roche diabetes. The other authors have no information to disclose.

Grants and fellowships supporting the writing of the paper

Sarah Flanagan is supported by a Sir Henry Dale Fellowship jointly funded by the Wellcome Trust and the Royal Society (105,636/Z/14/Z). The other authors are not supported by grants or fellowships.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
1.
Zurück zum Zitat Ahrén B. Autonomic regulation of islet hormone secretion--implications for health and disease. Diabetologia. 2000;43(4):393–410.PubMed Ahrén B. Autonomic regulation of islet hormone secretion--implications for health and disease. Diabetologia. 2000;43(4):393–410.PubMed
2.
Zurück zum Zitat Senniappan S, Shanti B, James C, Hussain K. Hyperinsulinaemic hypoglycaemia: genetic mechanisms, diagnosis and management. J Inherit Metab Dis. 2012;35(4):589–601.PubMed Senniappan S, Shanti B, James C, Hussain K. Hyperinsulinaemic hypoglycaemia: genetic mechanisms, diagnosis and management. J Inherit Metab Dis. 2012;35(4):589–601.PubMed
3.
Zurück zum Zitat Chinoy, A. et al. ‘Focal congenital hyperinsulinism as a cause for sudden infant death’. Pediatr Dev Pathol. 2019;22(1):65–69. Chinoy, A. et al. ‘Focal congenital hyperinsulinism as a cause for sudden infant death’. Pediatr Dev Pathol. 2019;22(1):65–69.
4.
Zurück zum Zitat Guyot A, Moreau F, Eberhard M, Gaulier JM, Paraf F. Congenital hyperinsulinism revealed by sudden infant death. Ann Pathol. 2017;37(5):429–32.PubMed Guyot A, Moreau F, Eberhard M, Gaulier JM, Paraf F. Congenital hyperinsulinism revealed by sudden infant death. Ann Pathol. 2017;37(5):429–32.PubMed
5.
Zurück zum Zitat Avatapalle HB, et al. Abnormal neurodevelopmental outcomes are common in children with transient congenital Hyperinsulinism. Front Endocrinol (Lausanne). 2013;4:60. Avatapalle HB, et al. Abnormal neurodevelopmental outcomes are common in children with transient congenital Hyperinsulinism. Front Endocrinol (Lausanne). 2013;4:60.
6.
Zurück zum Zitat Güemes M, Rahman SA, Hussain K. What is a normal blood glucose? Arch Dis Child. 2016;101:569–574. Güemes M, Rahman SA, Hussain K. What is a normal blood glucose? Arch Dis Child. 2016;101:569–574.
7.
Zurück zum Zitat Thornton PS, Stanley CA, de Leon DD, Harris D, Haymond MW, Hussain K, et al. Recommendations from the pediatric Endocrine Society for evaluation and Management of Persistent Hypoglycemia in neonates, infants, and children. J Pediatr. 2015;167(2):238–45.PubMed Thornton PS, Stanley CA, de Leon DD, Harris D, Haymond MW, Hussain K, et al. Recommendations from the pediatric Endocrine Society for evaluation and Management of Persistent Hypoglycemia in neonates, infants, and children. J Pediatr. 2015;167(2):238–45.PubMed
8.
Zurück zum Zitat Hussain K. Diagnosis and management of hyperinsulinaemic hypoglycaemia of infancy. Horm Res. 2008;69(1):2–13.PubMed Hussain K. Diagnosis and management of hyperinsulinaemic hypoglycaemia of infancy. Horm Res. 2008;69(1):2–13.PubMed
9.
Zurück zum Zitat Patel P, Charles L, Corbin J, et al. A unique allosteric insulin receptor monoclonal antibody that prevents hypoglycemia in the SUR-1-/- mouse model of KATP hyperinsulinism. MAbs. 2018;10(5):796–802. Patel P, Charles L, Corbin J, et al. A unique allosteric insulin receptor monoclonal antibody that prevents hypoglycemia in the SUR-1-/- mouse model of KATP hyperinsulinism. MAbs. 2018;10(5):796–802. 
10.
Zurück zum Zitat Arya VB, Mohammed Z, Blankenstein O, de Lonlay P, Hussain K. Hyperinsulinaemic hypoglycaemia. Horm Metab Res. 2014;46(3):157–70. Arya VB, Mohammed Z, Blankenstein O, de Lonlay P, Hussain K. Hyperinsulinaemic hypoglycaemia. Horm Metab Res. 2014;46(3):157–70.
11.
Zurück zum Zitat Vannucci RC, Vannucci SJ. Hypoglycemic brain injury. Semin Neonatol. 2001;6(2):147–55. Vannucci RC, Vannucci SJ. Hypoglycemic brain injury. Semin Neonatol. 2001;6(2):147–55.
12.
Zurück zum Zitat Hussain K, et al. An activating mutation of AKT2 and human hypoglycemia. Science. 2011;334(6055):474. Hussain K, et al. An activating mutation of AKT2 and human hypoglycemia. Science. 2011;334(6055):474.
13.
Zurück zum Zitat Staufner C, Lindner M, Dionisi-Vici C, Freisinger P, Dobbelaere D, Douillard C, et al. Adenosine kinase deficiency: expanding the clinical spectrum and evaluating therapeutic options. J Inherit Metab Dis. 2016;39(2):273–83. Staufner C, Lindner M, Dionisi-Vici C, Freisinger P, Dobbelaere D, Douillard C, et al. Adenosine kinase deficiency: expanding the clinical spectrum and evaluating therapeutic options. J Inherit Metab Dis. 2016;39(2):273–83.
14.
Zurück zum Zitat Gillis D, Krishnamohan A, Yaacov B, Shaag A, Jackman JE, Elpeleg O. TRMT10A dysfunction is associated with abnormalities in glucose homeostasis, short stature and microcephaly. J Med Genet. 2014;51(9):581–6. Gillis D, Krishnamohan A, Yaacov B, Shaag A, Jackman JE, Elpeleg O. TRMT10A dysfunction is associated with abnormalities in glucose homeostasis, short stature and microcephaly. J Med Genet. 2014;51(9):581–6.
15.
Zurück zum Zitat Senniappan S, Arya VB, Hussain K. The molecular mechanisms, diagnosis and management of congenital hyperinsulinism. Indian J Endocrinol Metab. 2013;17(1):19–30.PubMedPubMedCentral Senniappan S, Arya VB, Hussain K. The molecular mechanisms, diagnosis and management of congenital hyperinsulinism. Indian J Endocrinol Metab. 2013;17(1):19–30.PubMedPubMedCentral
16.
Zurück zum Zitat Inagaki N, et al. Reconstitution of IKATP: an inward rectifier subunit plus the sulfonylurea receptor. Science. 1995;270(5239):1166–70.PubMed Inagaki N, et al. Reconstitution of IKATP: an inward rectifier subunit plus the sulfonylurea receptor. Science. 1995;270(5239):1166–70.PubMed
17.
Zurück zum Zitat Kapoor RR, et al. Clinical and molecular characterisation of 300 patients with congenital hyperinsulinism. Eur J Endocrinol. 2013;168(4):557–64.PubMedPubMedCentral Kapoor RR, et al. Clinical and molecular characterisation of 300 patients with congenital hyperinsulinism. Eur J Endocrinol. 2013;168(4):557–64.PubMedPubMedCentral
18.
Zurück zum Zitat Thomas PM, Cote GJ, Wohllk N, Haddad B, Mathew PM, Rabl W, et al. Mutations in the sulfonylurea receptor gene in familial persistent hyperinsulinemic hypoglycemia of infancy. Science. 1995;268(5209):426–9.PubMed Thomas PM, Cote GJ, Wohllk N, Haddad B, Mathew PM, Rabl W, et al. Mutations in the sulfonylurea receptor gene in familial persistent hyperinsulinemic hypoglycemia of infancy. Science. 1995;268(5209):426–9.PubMed
19.
Zurück zum Zitat Thomas P, Ye Y, Lightner E. Mutation of the pancreatic islet inward rectifier Kir6.2 also leads to familial persistent hyperinsulinemic hypoglycemia of infancy. Hum Mol Genet. 1996;5(11):1809–12.PubMed Thomas P, Ye Y, Lightner E. Mutation of the pancreatic islet inward rectifier Kir6.2 also leads to familial persistent hyperinsulinemic hypoglycemia of infancy. Hum Mol Genet. 1996;5(11):1809–12.PubMed
20.
Zurück zum Zitat Taschenberger G, Mougey A, Shen S, Lester LB, LaFranchi S, Shyng SL. Identification of a familial hyperinsulinism-causing mutation in the sulfonylurea receptor 1 that prevents normal trafficking and function of KATP channels. J Biol Chem. 2002;277(19):17139–46.PubMed Taschenberger G, Mougey A, Shen S, Lester LB, LaFranchi S, Shyng SL. Identification of a familial hyperinsulinism-causing mutation in the sulfonylurea receptor 1 that prevents normal trafficking and function of KATP channels. J Biol Chem. 2002;277(19):17139–46.PubMed
21.
Zurück zum Zitat Huopio H, et al. K(ATP) channels and insulin secretion disorders. Am J Physiol Endocrinol Metab. 2002;283(2):E207–16.PubMed Huopio H, et al. K(ATP) channels and insulin secretion disorders. Am J Physiol Endocrinol Metab. 2002;283(2):E207–16.PubMed
22.
Zurück zum Zitat Huopio H, Reimann F, Ashfield R, Komulainen J, Lenko HL, Rahier J, et al. Dominantly inherited hyperinsulinism caused by a mutation in the sulfonylurea receptor type 1. J Clin Invest. 2000;106(7):897–906.PubMedPubMedCentral Huopio H, Reimann F, Ashfield R, Komulainen J, Lenko HL, Rahier J, et al. Dominantly inherited hyperinsulinism caused by a mutation in the sulfonylurea receptor type 1. J Clin Invest. 2000;106(7):897–906.PubMedPubMedCentral
23.
Zurück zum Zitat Huopio H, Otonkoski T, Vauhkonen I, Reimann F, Ashcroft FM, Laakso M. A new subtype of autosomal dominant diabetes attributable to a mutation in the gene for sulfonylurea receptor 1. Lancet. 2003;361(9354):301–7.PubMed Huopio H, Otonkoski T, Vauhkonen I, Reimann F, Ashcroft FM, Laakso M. A new subtype of autosomal dominant diabetes attributable to a mutation in the gene for sulfonylurea receptor 1. Lancet. 2003;361(9354):301–7.PubMed
24.
Zurück zum Zitat Flanagan SE, Kapoor RR, Banerjee I, Hall C, Smith VV, Hussain K, et al. Dominantly acting ABCC8 mutations in patients with medically unresponsive hyperinsulinaemic hypoglycaemia. Clin Genet. 2011;79(6):582–7.PubMedPubMedCentral Flanagan SE, Kapoor RR, Banerjee I, Hall C, Smith VV, Hussain K, et al. Dominantly acting ABCC8 mutations in patients with medically unresponsive hyperinsulinaemic hypoglycaemia. Clin Genet. 2011;79(6):582–7.PubMedPubMedCentral
25.
Zurück zum Zitat Pinney SE, MacMullen C, Becker S, Lin YW, Hanna C, Thornton P, et al. Clinical characteristics and biochemical mechanisms of congenital hyperinsulinism associated with dominant KATP channel mutations. J Clin Invest. 2008;118(8):2877–86.PubMedPubMedCentral Pinney SE, MacMullen C, Becker S, Lin YW, Hanna C, Thornton P, et al. Clinical characteristics and biochemical mechanisms of congenital hyperinsulinism associated with dominant KATP channel mutations. J Clin Invest. 2008;118(8):2877–86.PubMedPubMedCentral
26.
Zurück zum Zitat Kapoor RR, et al. Hyperinsulinaemic hypoglycaemia and diabetes mellitus due to dominant ABCC8/KCNJ11 mutations. Diabetologia. 2011;54(10):2575–83.PubMedPubMedCentral Kapoor RR, et al. Hyperinsulinaemic hypoglycaemia and diabetes mellitus due to dominant ABCC8/KCNJ11 mutations. Diabetologia. 2011;54(10):2575–83.PubMedPubMedCentral
27.
Zurück zum Zitat Stanley CA, et al. Hyperinsulinism and hyperammonemia in infants with regulatory mutations of the glutamate dehydrogenase gene. N Engl J Med. 1998;338(19):1352–7.PubMed Stanley CA, et al. Hyperinsulinism and hyperammonemia in infants with regulatory mutations of the glutamate dehydrogenase gene. N Engl J Med. 1998;338(19):1352–7.PubMed
28.
Zurück zum Zitat Stanley CA, Fang J, Kutyna K, Hsu BY, Ming JE, Glaser B, et al. Molecular basis and characterization of the hyperinsulinism/hyperammonemia syndrome: predominance of mutations in exons 11 and 12 of the glutamate dehydrogenase gene. HI/HA Contributing Investigators. Diabetes. 2000;49(4):667–73.PubMed Stanley CA, Fang J, Kutyna K, Hsu BY, Ming JE, Glaser B, et al. Molecular basis and characterization of the hyperinsulinism/hyperammonemia syndrome: predominance of mutations in exons 11 and 12 of the glutamate dehydrogenase gene. HI/HA Contributing Investigators. Diabetes. 2000;49(4):667–73.PubMed
29.
Zurück zum Zitat Kapoor RR, Flanagan SE, Fulton P, Chakrapani A, Chadefaux B, Ben-Omran T, et al. Hyperinsulinism-hyperammonaemia syndrome: novel mutations in the GLUD1 gene and genotype-phenotype correlations. Eur J Endocrinol. 2009;161(5):731–5.PubMedPubMedCentral Kapoor RR, Flanagan SE, Fulton P, Chakrapani A, Chadefaux B, Ben-Omran T, et al. Hyperinsulinism-hyperammonaemia syndrome: novel mutations in the GLUD1 gene and genotype-phenotype correlations. Eur J Endocrinol. 2009;161(5):731–5.PubMedPubMedCentral
30.
Zurück zum Zitat Glaser B, et al. Familial hyperinsulinism caused by an activating glucokinase mutation. N Engl J Med. 1998;338(4):226–30.PubMed Glaser B, et al. Familial hyperinsulinism caused by an activating glucokinase mutation. N Engl J Med. 1998;338(4):226–30.PubMed
31.
Zurück zum Zitat Christesen HB, et al. Activating glucokinase (GCK) mutations as a cause of medically responsive congenital hyperinsulinism: prevalence in children and characterisation of a novel GCK mutation. Eur J Endocrinol. 2008;159(1):27–34.PubMed Christesen HB, et al. Activating glucokinase (GCK) mutations as a cause of medically responsive congenital hyperinsulinism: prevalence in children and characterisation of a novel GCK mutation. Eur J Endocrinol. 2008;159(1):27–34.PubMed
32.
Zurück zum Zitat Cuesta-Munoz AL, et al. Severe persistent hyperinsulinemic hypoglycemia due to a de novo glucokinase mutation. Diabetes. 2004;53(8):2164–8.PubMed Cuesta-Munoz AL, et al. Severe persistent hyperinsulinemic hypoglycemia due to a de novo glucokinase mutation. Diabetes. 2004;53(8):2164–8.PubMed
33.
Zurück zum Zitat Christesen HB, Jacobsen BB, Odili S, Buettger C, Cuesta-Munoz A, Hansen T, et al. The second activating glucokinase mutation (A456V): implications for glucose homeostasis and diabetes therapy. Diabetes. 2002;51(4):1240–6.PubMed Christesen HB, Jacobsen BB, Odili S, Buettger C, Cuesta-Munoz A, Hansen T, et al. The second activating glucokinase mutation (A456V): implications for glucose homeostasis and diabetes therapy. Diabetes. 2002;51(4):1240–6.PubMed
34.
Zurück zum Zitat Heslegrave AJ, et al. Leucine-sensitive hyperinsulinaemic hypoglycaemia in patients with loss of function mutations in 3-Hydroxyacyl-CoA dehydrogenase. Orphanet J Rare Dis. 2012;7:25.PubMedPubMedCentral Heslegrave AJ, et al. Leucine-sensitive hyperinsulinaemic hypoglycaemia in patients with loss of function mutations in 3-Hydroxyacyl-CoA dehydrogenase. Orphanet J Rare Dis. 2012;7:25.PubMedPubMedCentral
35.
Zurück zum Zitat Filling C, Keller B, Hirschberg D, Marschall HU, Jörnvall H, Bennett MJ, et al. Role of short-chain hydroxyacyl CoA dehydrogenases in SCHAD deficiency. Biochem Biophys Res Commun. 2008;368(1):6–11.PubMed Filling C, Keller B, Hirschberg D, Marschall HU, Jörnvall H, Bennett MJ, et al. Role of short-chain hydroxyacyl CoA dehydrogenases in SCHAD deficiency. Biochem Biophys Res Commun. 2008;368(1):6–11.PubMed
36.
Zurück zum Zitat Clayton PT, et al. Hyperinsulinism in short-chain L-3-hydroxyacyl-CoA dehydrogenase deficiency reveals the importance of beta-oxidation in insulin secretion. J Clin Invest. 2001;108(3):457–65.PubMedPubMedCentral Clayton PT, et al. Hyperinsulinism in short-chain L-3-hydroxyacyl-CoA dehydrogenase deficiency reveals the importance of beta-oxidation in insulin secretion. J Clin Invest. 2001;108(3):457–65.PubMedPubMedCentral
37.
Zurück zum Zitat Molven A, Matre GE, Duran M, Wanders RJ, Rishaug U, Njølstad PR, et al. Familial hyperinsulinemic hypoglycemia caused by a defect in the SCHAD enzyme of mitochondrial fatty acid oxidation. Diabetes. 2004;53(1):221–7.PubMed Molven A, Matre GE, Duran M, Wanders RJ, Rishaug U, Njølstad PR, et al. Familial hyperinsulinemic hypoglycemia caused by a defect in the SCHAD enzyme of mitochondrial fatty acid oxidation. Diabetes. 2004;53(1):221–7.PubMed
38.
Zurück zum Zitat Kapoor RR, James C, Flanagan SE, Ellard S, Eaton S, Hussain K. 3-Hydroxyacyl-coenzyme a dehydrogenase deficiency and hyperinsulinemic hypoglycemia: characterization of a novel mutation and severe dietary protein sensitivity. J Clin Endocrinol Metab. 2009;94(7):2221–5.PubMed Kapoor RR, James C, Flanagan SE, Ellard S, Eaton S, Hussain K. 3-Hydroxyacyl-coenzyme a dehydrogenase deficiency and hyperinsulinemic hypoglycemia: characterization of a novel mutation and severe dietary protein sensitivity. J Clin Endocrinol Metab. 2009;94(7):2221–5.PubMed
39.
Zurück zum Zitat Flanagan SE, et al. Genome-wide homozygosity analysis reveals HADH mutations as a common cause of diazoxide-responsive hyperinsulinemic-hypoglycemia in consanguineous pedigrees. J Clin Endocrinol Metab. 2011;96(3):E498–502.PubMedPubMedCentral Flanagan SE, et al. Genome-wide homozygosity analysis reveals HADH mutations as a common cause of diazoxide-responsive hyperinsulinemic-hypoglycemia in consanguineous pedigrees. J Clin Endocrinol Metab. 2011;96(3):E498–502.PubMedPubMedCentral
40.
Zurück zum Zitat Colclough K, Bellanne-Chantelot C, Saint-Martin C, Flanagan SE, Ellard S. Mutations in the genes encoding the transcription factors hepatocyte nuclear factor 1 alpha and 4 alpha in maturity-onset diabetes of the young and hyperinsulinemic hypoglycemia. Hum Mutat. 2013;34(5):669–85.PubMed Colclough K, Bellanne-Chantelot C, Saint-Martin C, Flanagan SE, Ellard S. Mutations in the genes encoding the transcription factors hepatocyte nuclear factor 1 alpha and 4 alpha in maturity-onset diabetes of the young and hyperinsulinemic hypoglycemia. Hum Mutat. 2013;34(5):669–85.PubMed
41.
Zurück zum Zitat Pearson ER, Boj SF, Steele AM, Barrett T, Stals K, Shield JP, et al. Macrosomia and hyperinsulinaemic hypoglycaemia in patients with heterozygous mutations in the HNF4A gene. PLoS Med. 2007;4(4):e118.PubMedPubMedCentral Pearson ER, Boj SF, Steele AM, Barrett T, Stals K, Shield JP, et al. Macrosomia and hyperinsulinaemic hypoglycaemia in patients with heterozygous mutations in the HNF4A gene. PLoS Med. 2007;4(4):e118.PubMedPubMedCentral
42.
Zurück zum Zitat Kapoor RR, Locke J, Colclough K, Wales J, Conn JJ, Hattersley AT, et al. Persistent hyperinsulinemic hypoglycemia and maturity-onset diabetes of the young due to heterozygous HNF4A mutations. Diabetes. 2008;57(6):1659–63.PubMed Kapoor RR, Locke J, Colclough K, Wales J, Conn JJ, Hattersley AT, et al. Persistent hyperinsulinemic hypoglycemia and maturity-onset diabetes of the young due to heterozygous HNF4A mutations. Diabetes. 2008;57(6):1659–63.PubMed
43.
Zurück zum Zitat Flanagan SE, et al. Diazoxide-responsive hyperinsulinemic hypoglycemia caused by HNF4A gene mutations. Eur J Endocrinol. 2010;162(5):987–92.PubMedPubMedCentral Flanagan SE, et al. Diazoxide-responsive hyperinsulinemic hypoglycemia caused by HNF4A gene mutations. Eur J Endocrinol. 2010;162(5):987–92.PubMedPubMedCentral
44.
Zurück zum Zitat McGlacken-Byrne SM, et al. The evolving course of HNF4A hyperinsulinaemic hypoglycaemia--a case series. Diabet Med. 2014;31(1):e1–5.PubMed McGlacken-Byrne SM, et al. The evolving course of HNF4A hyperinsulinaemic hypoglycaemia--a case series. Diabet Med. 2014;31(1):e1–5.PubMed
45.
Zurück zum Zitat Stanescu DE, Hughes N, Kaplan B, Stanley CA, de León DD. Novel presentations of congenital hyperinsulinism due to mutations in the MODY genes: HNF1A and HNF4A. J Clin Endocrinol Metab. 2012;97(10):E2026–30.PubMedPubMedCentral Stanescu DE, Hughes N, Kaplan B, Stanley CA, de León DD. Novel presentations of congenital hyperinsulinism due to mutations in the MODY genes: HNF1A and HNF4A. J Clin Endocrinol Metab. 2012;97(10):E2026–30.PubMedPubMedCentral
46.
Zurück zum Zitat Hamilton AJ, Bingham C, McDonald T, Cook PR, Caswell RC, Weedon MN, et al. The HNF4A R76W mutation causes atypical dominant Fanconi syndrome in addition to a beta cell phenotype. J Med Genet. 2014;51(3):165–9.PubMed Hamilton AJ, Bingham C, McDonald T, Cook PR, Caswell RC, Weedon MN, et al. The HNF4A R76W mutation causes atypical dominant Fanconi syndrome in addition to a beta cell phenotype. J Med Genet. 2014;51(3):165–9.PubMed
47.
Zurück zum Zitat Numakura C, Hashimoto Y, Daitsu T, Hayasaka K, Mitsui T, Yorifuji T. Two patients with HNF4A-related congenital hyperinsulinism and renal tubular dysfunction: a clinical variation which includes transient hepatic dysfunction. Diabetes Res Clin Pract. 2015;108(3):e53–5.PubMed Numakura C, Hashimoto Y, Daitsu T, Hayasaka K, Mitsui T, Yorifuji T. Two patients with HNF4A-related congenital hyperinsulinism and renal tubular dysfunction: a clinical variation which includes transient hepatic dysfunction. Diabetes Res Clin Pract. 2015;108(3):e53–5.PubMed
48.
Zurück zum Zitat Walsh SB, Unwin R, Kleta R, van't Hoff W, Bass P, Hussain K, et al. Fainting Fanconi syndrome clarified by proxy: a case report. BMC Nephrol. 2017;18(1):230.PubMedPubMedCentral Walsh SB, Unwin R, Kleta R, van't Hoff W, Bass P, Hussain K, et al. Fainting Fanconi syndrome clarified by proxy: a case report. BMC Nephrol. 2017;18(1):230.PubMedPubMedCentral
49.
Zurück zum Zitat Rozenkova K, Malikova J, Nessa A, Dusatkova L, Bjørkhaug L, Obermannova B, et al. High incidence of heterozygous ABCC8 and HNF1A mutations in Czech patients with congenital Hyperinsulinism. J Clin Endocrinol Metab. 2015;100(12):E1540–9.PubMed Rozenkova K, Malikova J, Nessa A, Dusatkova L, Bjørkhaug L, Obermannova B, et al. High incidence of heterozygous ABCC8 and HNF1A mutations in Czech patients with congenital Hyperinsulinism. J Clin Endocrinol Metab. 2015;100(12):E1540–9.PubMed
50.
Zurück zum Zitat Meissner T, Otonkoski T, Feneberg R, Beinbrech B, Apostolidou S, Sipilä I, et al. Exercise induced hypoglycaemic hyperinsulinism. Arch Dis Child. 2001;84(3):254–7.PubMedPubMedCentral Meissner T, Otonkoski T, Feneberg R, Beinbrech B, Apostolidou S, Sipilä I, et al. Exercise induced hypoglycaemic hyperinsulinism. Arch Dis Child. 2001;84(3):254–7.PubMedPubMedCentral
51.
Zurück zum Zitat Otonkoski T, Jiao H, Kaminen-Ahola N, Tapia-Paez I, Ullah MS, Parton LE, et al. Physical exercise-induced hypoglycemia caused by failed silencing of monocarboxylate transporter 1 in pancreatic beta cells. Am J Hum Genet. 2007;81(3):467–74.PubMedPubMedCentral Otonkoski T, Jiao H, Kaminen-Ahola N, Tapia-Paez I, Ullah MS, Parton LE, et al. Physical exercise-induced hypoglycemia caused by failed silencing of monocarboxylate transporter 1 in pancreatic beta cells. Am J Hum Genet. 2007;81(3):467–74.PubMedPubMedCentral
52.
Zurück zum Zitat Meissner T, Friedmann B, Okun JG, Schwab MA, Otonkoski T, Bauer T, et al. Massive insulin secretion in response to anaerobic exercise in exercise-induced hyperinsulinism. Horm Metab Res. 2005;37(11):690–4.PubMed Meissner T, Friedmann B, Okun JG, Schwab MA, Otonkoski T, Bauer T, et al. Massive insulin secretion in response to anaerobic exercise in exercise-induced hyperinsulinism. Horm Metab Res. 2005;37(11):690–4.PubMed
53.
Zurück zum Zitat Fleury C, et al. Uncoupling protein-2: a novel gene linked to obesity and hyperinsulinemia. Nat Genet. 1997;15(3):269–72.PubMed Fleury C, et al. Uncoupling protein-2: a novel gene linked to obesity and hyperinsulinemia. Nat Genet. 1997;15(3):269–72.PubMed
54.
Zurück zum Zitat González-Barroso MM, et al. Mutations in UCP2 in congenital hyperinsulinism reveal a role for regulation of insulin secretion. PLoS One. 2008;3(12):e3850.PubMedPubMedCentral González-Barroso MM, et al. Mutations in UCP2 in congenital hyperinsulinism reveal a role for regulation of insulin secretion. PLoS One. 2008;3(12):e3850.PubMedPubMedCentral
55.
Zurück zum Zitat Ferrara CT, Boodhansingh KE, Paradies E, Fiermonte G, Steinkrauss LJ, Topor LS, et al. Novel hypoglycemia phenotype in congenital Hyperinsulinism due to dominant mutations of uncoupling protein 2. J Clin Endocrinol Metab. 2017;102(3):942–9.PubMed Ferrara CT, Boodhansingh KE, Paradies E, Fiermonte G, Steinkrauss LJ, Topor LS, et al. Novel hypoglycemia phenotype in congenital Hyperinsulinism due to dominant mutations of uncoupling protein 2. J Clin Endocrinol Metab. 2017;102(3):942–9.PubMed
56.
Zurück zum Zitat Laver TW, Weedon MN, Caswell R, Hussain K, Ellard S, Flanagan SE. Analysis of large-scale sequencing cohorts does not support the role of variants in UCP2 as a cause of hyperinsulinaemic hypoglycaemia. Hum Mutat. 2017;38(10):1442–4.PubMedPubMedCentral Laver TW, Weedon MN, Caswell R, Hussain K, Ellard S, Flanagan SE. Analysis of large-scale sequencing cohorts does not support the role of variants in UCP2 as a cause of hyperinsulinaemic hypoglycaemia. Hum Mutat. 2017;38(10):1442–4.PubMedPubMedCentral
57.
Zurück zum Zitat Pinney SE, Ganapathy K, Bradfield J, Stokes D, Sasson A, Mackiewicz K, et al. Dominant form of congenital hyperinsulinism maps to HK1 region on 10q. Horm Res Paediatr. 2013;80(1):18–27.PubMed Pinney SE, Ganapathy K, Bradfield J, Stokes D, Sasson A, Mackiewicz K, et al. Dominant form of congenital hyperinsulinism maps to HK1 region on 10q. Horm Res Paediatr. 2013;80(1):18–27.PubMed
58.
Zurück zum Zitat Henquin JC, Sempoux C, Marchandise J, Godecharles S, Guiot Y, Nenquin M, et al. Congenital hyperinsulinism caused by hexokinase I expression or glucokinase-activating mutation in a subset of β-cells. Diabetes. 2013;62(5):1689–96.PubMedPubMedCentral Henquin JC, Sempoux C, Marchandise J, Godecharles S, Guiot Y, Nenquin M, et al. Congenital hyperinsulinism caused by hexokinase I expression or glucokinase-activating mutation in a subset of β-cells. Diabetes. 2013;62(5):1689–96.PubMedPubMedCentral
59.
60.
Zurück zum Zitat Cabezas OR, Flanagan SE, Stanescu H, García-Martínez E, Caswell R, Lango-Allen H, et al. Polycystic kidney disease with Hyperinsulinemic hypoglycemia caused by a promoter mutation in Phosphomannomutase 2. J Am Soc Nephrol. 2017;28(8):2529–39.PubMedPubMedCentral Cabezas OR, Flanagan SE, Stanescu H, García-Martínez E, Caswell R, Lango-Allen H, et al. Polycystic kidney disease with Hyperinsulinemic hypoglycemia caused by a promoter mutation in Phosphomannomutase 2. J Am Soc Nephrol. 2017;28(8):2529–39.PubMedPubMedCentral
61.
Zurück zum Zitat Giri D, Vignola ML, Gualtieri A, Scagliotti V, McNamara P, Peak M, et al. Novel FOXA2 mutation causes Hyperinsulinism, hypopituitarism with craniofacial and endoderm-derived organ abnormalities. Hum Mol Genet. 2017;26(22):4315–26.PubMed Giri D, Vignola ML, Gualtieri A, Scagliotti V, McNamara P, Peak M, et al. Novel FOXA2 mutation causes Hyperinsulinism, hypopituitarism with craniofacial and endoderm-derived organ abnormalities. Hum Mol Genet. 2017;26(22):4315–26.PubMed
62.
Zurück zum Zitat Vajravelu ME, et al. Congenital Hyperinsulinism and hypopituitarism attributable to a mutation in FOXA2. J Clin Endocrinol Metab. 2018;103(3):1042–7.PubMedPubMedCentral Vajravelu ME, et al. Congenital Hyperinsulinism and hypopituitarism attributable to a mutation in FOXA2. J Clin Endocrinol Metab. 2018;103(3):1042–7.PubMedPubMedCentral
63.
Zurück zum Zitat Flanagan SE, Vairo F, Johnson MB, Caswell R, Laver TW, Lango Allen H, et al. A CACNA1D mutation in a patient with persistent hyperinsulinaemic hypoglycaemia, heart defects, and severe hypotonia. Pediatr Diabetes. 2017;18(4):320–3.PubMedPubMedCentral Flanagan SE, Vairo F, Johnson MB, Caswell R, Laver TW, Lango Allen H, et al. A CACNA1D mutation in a patient with persistent hyperinsulinaemic hypoglycaemia, heart defects, and severe hypotonia. Pediatr Diabetes. 2017;18(4):320–3.PubMedPubMedCentral
64.
Zurück zum Zitat Gregory LC, Ferreira CB, Young-Baird SK, Williams HJ, Harakalova M, van Haaften G, et al. Impaired EIF2S3 function associated with a novel phenotype of X-linked hypopituitarism with glucose dysregulation. EBioMedicine. 2019;42:470–80.PubMedPubMedCentral Gregory LC, Ferreira CB, Young-Baird SK, Williams HJ, Harakalova M, van Haaften G, et al. Impaired EIF2S3 function associated with a novel phenotype of X-linked hypopituitarism with glucose dysregulation. EBioMedicine. 2019;42:470–80.PubMedPubMedCentral
65.
Zurück zum Zitat Bufler P, Ehringhaus C, Koletzko S. Dumping syndrome: a common problem following Nissen fundoplication in young children. Pediatr Surg Int. 2001;17(5–6):351–5.PubMed Bufler P, Ehringhaus C, Koletzko S. Dumping syndrome: a common problem following Nissen fundoplication in young children. Pediatr Surg Int. 2001;17(5–6):351–5.PubMed
66.
Zurück zum Zitat Foster-Schubert KE. Hypoglycemia complicating bariatric surgery: incidence and mechanisms. Curr Opin Endocrinol Diabetes Obes. 2011;18(2):129–33.PubMedPubMedCentral Foster-Schubert KE. Hypoglycemia complicating bariatric surgery: incidence and mechanisms. Curr Opin Endocrinol Diabetes Obes. 2011;18(2):129–33.PubMedPubMedCentral
67.
Zurück zum Zitat Palladino AA, et al. Increased glucagon-like peptide-1 secretion and postprandial hypoglycemia in children after Nissen fundoplication. J Clin Endocrinol Metab. 2009;94(1):39–44.PubMed Palladino AA, et al. Increased glucagon-like peptide-1 secretion and postprandial hypoglycemia in children after Nissen fundoplication. J Clin Endocrinol Metab. 2009;94(1):39–44.PubMed
68.
Zurück zum Zitat Hirata Y. Insulin autoimmune syndrome. Nihon Rinsho. 1973;31(7):2227–31.PubMed Hirata Y. Insulin autoimmune syndrome. Nihon Rinsho. 1973;31(7):2227–31.PubMed
69.
Zurück zum Zitat Shin JJ, Gorden P, Libutti SK. Insulinoma: pathophysiology, localization and management. Future Oncol. 2010;6(2):229–37.PubMedPubMedCentral Shin JJ, Gorden P, Libutti SK. Insulinoma: pathophysiology, localization and management. Future Oncol. 2010;6(2):229–37.PubMedPubMedCentral
70.
Zurück zum Zitat Ozon A, Demirbilek H, Ertugrul A, Unal S, Gumruk F, Kandemir N. Anemia and neutropenic fever with high dose diazoxide treatment in a case with hyperinsulinism due to Munchausen by proxy. J Pediatr Endocrinol Metab. 2010;23(7):719–23.PubMed Ozon A, Demirbilek H, Ertugrul A, Unal S, Gumruk F, Kandemir N. Anemia and neutropenic fever with high dose diazoxide treatment in a case with hyperinsulinism due to Munchausen by proxy. J Pediatr Endocrinol Metab. 2010;23(7):719–23.PubMed
71.
Zurück zum Zitat Toda N, Ihara K, Kojima-Ishii K, Ochiai M, Ohkubo K, Kawamoto Y, et al. Hyperinsulinemic hypoglycemia in Beckwith-Wiedemann, Sotos, and kabuki syndromes: a nationwide survey in Japan. Am J Med Genet A. 2017;173(2):360–7.PubMed Toda N, Ihara K, Kojima-Ishii K, Ochiai M, Ohkubo K, Kawamoto Y, et al. Hyperinsulinemic hypoglycemia in Beckwith-Wiedemann, Sotos, and kabuki syndromes: a nationwide survey in Japan. Am J Med Genet A. 2017;173(2):360–7.PubMed
72.
Zurück zum Zitat Henquin JC, Nenquin M, Sempoux C, Guiot Y, Bellanné-Chantelot C, Otonkoski T, et al. In vitro insulin secretion by pancreatic tissue from infants with diazoxide-resistant congenital hyperinsulinism deviates from model predictions. J Clin Invest. 2011;121(10):3932–42.PubMedPubMedCentral Henquin JC, Nenquin M, Sempoux C, Guiot Y, Bellanné-Chantelot C, Otonkoski T, et al. In vitro insulin secretion by pancreatic tissue from infants with diazoxide-resistant congenital hyperinsulinism deviates from model predictions. J Clin Invest. 2011;121(10):3932–42.PubMedPubMedCentral
73.
Zurück zum Zitat Sempoux C, et al. Morphological Mosaicism of the pancreatic islets: a novel Anatomopathological form of persistent Hyperinsulinemic hypoglycemia of infancy. J Clin Endocrinol Metab. 2011;96(12):3785–93.PubMed Sempoux C, et al. Morphological Mosaicism of the pancreatic islets: a novel Anatomopathological form of persistent Hyperinsulinemic hypoglycemia of infancy. J Clin Endocrinol Metab. 2011;96(12):3785–93.PubMed
74.
Zurück zum Zitat Damaj L, le Lorch M, Verkarre V, Werl C, Hubert L, Nihoul-Fékété C, et al. Chromosome 11p15 paternal isodisomy in focal forms of neonatal hyperinsulinism. J Clin Endocrinol Metab. 2008;93(12):4941–7.PubMed Damaj L, le Lorch M, Verkarre V, Werl C, Hubert L, Nihoul-Fékété C, et al. Chromosome 11p15 paternal isodisomy in focal forms of neonatal hyperinsulinism. J Clin Endocrinol Metab. 2008;93(12):4941–7.PubMed
75.
Zurück zum Zitat Rahier J, Fält K, Müntefering H, Becker K, Gepts W, Falkmer S. The basic structural lesion of persistent neonatal hypoglycaemia with hyperinsulinism: deficiency of pancreatic D cells or hyperactivity of B cells? Diabetologia. 1984;26(4):282–9.PubMed Rahier J, Fält K, Müntefering H, Becker K, Gepts W, Falkmer S. The basic structural lesion of persistent neonatal hypoglycaemia with hyperinsulinism: deficiency of pancreatic D cells or hyperactivity of B cells? Diabetologia. 1984;26(4):282–9.PubMed
76.
Zurück zum Zitat Rahier J, Guiot Y, Sempoux C. Persistent hyperinsulinaemic hypoglycaemia of infancy: a heterogeneous syndrome unrelated to nesidioblastosis. Arch Dis Child Fetal Neonatal Ed. 2000;82(2):F108–12.PubMedPubMedCentral Rahier J, Guiot Y, Sempoux C. Persistent hyperinsulinaemic hypoglycaemia of infancy: a heterogeneous syndrome unrelated to nesidioblastosis. Arch Dis Child Fetal Neonatal Ed. 2000;82(2):F108–12.PubMedPubMedCentral
77.
Zurück zum Zitat Goossens AGW. Saudubray JM, Bonnefont JP, Nihoul-Fekete, Heitz PU, Klöppel G., diffuse and focal nesidioblastosis. A clinicopathological study of 24 patients with persistent neonatal hyperinsulinemic hypoglycemia. Am J Surg Pathol. 1989;3(9):766–55. Goossens AGW. Saudubray JM, Bonnefont JP, Nihoul-Fekete, Heitz PU, Klöppel G., diffuse and focal nesidioblastosis. A clinicopathological study of 24 patients with persistent neonatal hyperinsulinemic hypoglycemia. Am J Surg Pathol. 1989;3(9):766–55.
78.
Zurück zum Zitat Sempoux C, et al. Neonatal hyperinsulinemic hypoglycemia: heterogeneity of the syndrome and keys for differential diagnosis. J Clin Endocrinol Metab. 1998;83(5):1455–61.PubMed Sempoux C, et al. Neonatal hyperinsulinemic hypoglycemia: heterogeneity of the syndrome and keys for differential diagnosis. J Clin Endocrinol Metab. 1998;83(5):1455–61.PubMed
79.
Zurück zum Zitat Rahier J, et al. Partial or near-total pancreatectomy for persistent neonatal hyperinsulinaemic hypoglycaemia: the pathologist's role. Histopathology. 1998;32(1):15–9.PubMed Rahier J, et al. Partial or near-total pancreatectomy for persistent neonatal hyperinsulinaemic hypoglycaemia: the pathologist's role. Histopathology. 1998;32(1):15–9.PubMed
80.
Zurück zum Zitat Otonkoski T, et al. Noninvasive diagnosis of focal Hyperinsulinism of infancy with [18F]-DOPA positron emission tomography. Diabetes. 2006;55(1):13–8.PubMed Otonkoski T, et al. Noninvasive diagnosis of focal Hyperinsulinism of infancy with [18F]-DOPA positron emission tomography. Diabetes. 2006;55(1):13–8.PubMed
81.
Zurück zum Zitat Sempoux C, Guiot Y, Jaubert F, Rahier J. Focal and diffuse forms of congenital hyperinsulinism: the keys for differential diagnosis. Endocr Pathol. 2004;15(3):241–6.PubMed Sempoux C, Guiot Y, Jaubert F, Rahier J. Focal and diffuse forms of congenital hyperinsulinism: the keys for differential diagnosis. Endocr Pathol. 2004;15(3):241–6.PubMed
82.
Zurück zum Zitat Hussain K, et al. An ABCC8 gene mutation and mosaic uniparental isodisomy resulting in atypical diffuse congenital hyperinsulinism. Diabetes. 2008;57(1):259–63.PubMed Hussain K, et al. An ABCC8 gene mutation and mosaic uniparental isodisomy resulting in atypical diffuse congenital hyperinsulinism. Diabetes. 2008;57(1):259–63.PubMed
83.
Zurück zum Zitat Henquin JC, Sempoux C, Marchandise J, Godecharles S, Guiot Y, Nenquin M, et al. Congenital hyperinsulinism caused by hexokinase I expression or glucokinase-activating mutation in a subset of beta-cells. Diabetes. 2013;62(5):1689–96.PubMedPubMedCentral Henquin JC, Sempoux C, Marchandise J, Godecharles S, Guiot Y, Nenquin M, et al. Congenital hyperinsulinism caused by hexokinase I expression or glucokinase-activating mutation in a subset of beta-cells. Diabetes. 2013;62(5):1689–96.PubMedPubMedCentral
84.
Zurück zum Zitat Shi Y, Avatapalle HB, Skae MS, Padidela R, Newbould M, Rigby L, et al. Increased plasma Incretin concentrations identifies a subset of patients with persistent congenital Hyperinsulinism without KATP Channel gene defects. J Pediatr. 2015;166(1):191–4.PubMed Shi Y, Avatapalle HB, Skae MS, Padidela R, Newbould M, Rigby L, et al. Increased plasma Incretin concentrations identifies a subset of patients with persistent congenital Hyperinsulinism without KATP Channel gene defects. J Pediatr. 2015;166(1):191–4.PubMed
85.
Zurück zum Zitat Hussain K. Investigations for neonatal hypoglycaemia. Clin Biochem. 2011;44(7):465–6.PubMed Hussain K. Investigations for neonatal hypoglycaemia. Clin Biochem. 2011;44(7):465–6.PubMed
86.
Zurück zum Zitat Aynsley-Green A, Hussain K, Hall J, Saudubray JM, Nihoul-Fékété C, de Lonlay-Debeney P, et al. Practical management of hyperinsulinism in infancy. Arch Dis Child Fetal Neonatal Ed. 2000;82(2):F98–F107.PubMedPubMedCentral Aynsley-Green A, Hussain K, Hall J, Saudubray JM, Nihoul-Fékété C, de Lonlay-Debeney P, et al. Practical management of hyperinsulinism in infancy. Arch Dis Child Fetal Neonatal Ed. 2000;82(2):F98–F107.PubMedPubMedCentral
87.
Zurück zum Zitat Yorifuji T, Horikawa R, Hasegawa T, Adachi M, Soneda S, Minagawa M, et al. Clinical practice guidelines for congenital hyperinsulinism. Clin Pediatr Endocrinol. 2017;26(3):127–52.PubMedPubMedCentral Yorifuji T, Horikawa R, Hasegawa T, Adachi M, Soneda S, Minagawa M, et al. Clinical practice guidelines for congenital hyperinsulinism. Clin Pediatr Endocrinol. 2017;26(3):127–52.PubMedPubMedCentral
88.
Zurück zum Zitat Palladino AA, Bennett MJ, Stanley CA. Hyperinsulinism in infancy and childhood: when an insulin level is not always enough. Clin Chem. 2008;54(2):256–63.PubMed Palladino AA, Bennett MJ, Stanley CA. Hyperinsulinism in infancy and childhood: when an insulin level is not always enough. Clin Chem. 2008;54(2):256–63.PubMed
89.
Zurück zum Zitat Al-Otaibi H, et al. Biochemical studies in patients with hyperinsulinaemic hypoglycaemia. Eur J Pediatr. 2013;172(11):1435–40.PubMed Al-Otaibi H, et al. Biochemical studies in patients with hyperinsulinaemic hypoglycaemia. Eur J Pediatr. 2013;172(11):1435–40.PubMed
90.
Zurück zum Zitat Ferrara C, et al. Biomarkers of insulin for the diagnosis of Hyperinsulinemic hypoglycemia in infants and children. J Pediatr. 2016;168:212–9. Ferrara C, et al. Biomarkers of insulin for the diagnosis of Hyperinsulinemic hypoglycemia in infants and children. J Pediatr. 2016;168:212–9.
91.
Zurück zum Zitat Brun JF, Fédou C, Bouix O, Raynaud E, Orsetti A. Evaluation of a standardized hyperglucidic breakfast test in postprandial reactive hypoglycaemia. Diabetologia. 1995;38(4):494–501.PubMed Brun JF, Fédou C, Bouix O, Raynaud E, Orsetti A. Evaluation of a standardized hyperglucidic breakfast test in postprandial reactive hypoglycaemia. Diabetologia. 1995;38(4):494–501.PubMed
92.
Zurück zum Zitat Otonkoski T, Kaminen N, Ustinov J, Lapatto R, Meissner T, Mayatepek E, et al. Physical exercise-induced hyperinsulinemic hypoglycemia is an autosomal-dominant trait characterized by abnormal pyruvate-induced insulin release. Diabetes. 2003;52(1):199–204.PubMed Otonkoski T, Kaminen N, Ustinov J, Lapatto R, Meissner T, Mayatepek E, et al. Physical exercise-induced hyperinsulinemic hypoglycemia is an autosomal-dominant trait characterized by abnormal pyruvate-induced insulin release. Diabetes. 2003;52(1):199–204.PubMed
93.
Zurück zum Zitat Alsaffar H, et al. Continuous flash glucose monitoring in children with congenital Hyperinsulinism; first report on accuracy and patient experience. Int J Pediatr Endocrinol. 2018;2018:3.PubMedPubMedCentral Alsaffar H, et al. Continuous flash glucose monitoring in children with congenital Hyperinsulinism; first report on accuracy and patient experience. Int J Pediatr Endocrinol. 2018;2018:3.PubMedPubMedCentral
94.
Zurück zum Zitat Meintjes M, Endozo R, Dickson J, Erlandsson K, Hussain K, Townsend C, et al. 18F-DOPA PET and enhanced CT imaging for congenital hyperinsulinism: initial UK experience from a technologist's perspective. Nucl Med Commun. 2013;34(6):601–8.PubMed Meintjes M, Endozo R, Dickson J, Erlandsson K, Hussain K, Townsend C, et al. 18F-DOPA PET and enhanced CT imaging for congenital hyperinsulinism: initial UK experience from a technologist's perspective. Nucl Med Commun. 2013;34(6):601–8.PubMed
95.
Zurück zum Zitat Lord K, et al. Clinical presentation and management of children with diffuse and focal hyperinsulinism: a review of 223 cases. J Clin Endocrinol Metab. 2013;98(11):E1786–9.PubMedPubMedCentral Lord K, et al. Clinical presentation and management of children with diffuse and focal hyperinsulinism: a review of 223 cases. J Clin Endocrinol Metab. 2013;98(11):E1786–9.PubMedPubMedCentral
96.
Zurück zum Zitat Blomberg BA, et al. The value of radiologic interventions and (18)F-DOPA PET in diagnosing and localizing focal congenital hyperinsulinism: systematic review and meta-analysis. Mol Imaging Biol. 2013;15(1):97–105.PubMed Blomberg BA, et al. The value of radiologic interventions and (18)F-DOPA PET in diagnosing and localizing focal congenital hyperinsulinism: systematic review and meta-analysis. Mol Imaging Biol. 2013;15(1):97–105.PubMed
97.
Zurück zum Zitat Ismail D, Kapoor RR, Smith VV, Ashworth M, Blankenstein O, Pierro A, et al. The heterogeneity of focal forms of congenital hyperinsulinism. J Clin Endocrinol Metab. 2012;97(1):E94–9.PubMed Ismail D, Kapoor RR, Smith VV, Ashworth M, Blankenstein O, Pierro A, et al. The heterogeneity of focal forms of congenital hyperinsulinism. J Clin Endocrinol Metab. 2012;97(1):E94–9.PubMed
98.
Zurück zum Zitat Hardy OT, et al. Accuracy of [18F]fluorodopa positron emission tomography for diagnosing and localizing focal congenital hyperinsulinism. J Clin Endocrinol Metab. 2007;92(12):4706–11.PubMed Hardy OT, et al. Accuracy of [18F]fluorodopa positron emission tomography for diagnosing and localizing focal congenital hyperinsulinism. J Clin Endocrinol Metab. 2007;92(12):4706–11.PubMed
99.
Zurück zum Zitat Banerjee I, Avatapalle B, Padidela R, Stevens A, Cosgrove KE, Clayton PE, et al. Integrating genetic and imaging investigations into the clinical management of congenital hyperinsulinism. Clin Endocrinol. 2013;78(6):803–13. Banerjee I, Avatapalle B, Padidela R, Stevens A, Cosgrove KE, Clayton PE, et al. Integrating genetic and imaging investigations into the clinical management of congenital hyperinsulinism. Clin Endocrinol. 2013;78(6):803–13.
100.
Zurück zum Zitat Ribeiro MJ, Boddaert N, Delzescaux T, Valayannopoulos V, Bellanné-Chantelot C, Jaubert F, et al. Functional imaging of the pancreas: the role of [18F]fluoro-L-DOPA PET in the diagnosis of hyperinsulinism of infancy. Endocr Dev. 2007;12:55–66.PubMed Ribeiro MJ, Boddaert N, Delzescaux T, Valayannopoulos V, Bellanné-Chantelot C, Jaubert F, et al. Functional imaging of the pancreas: the role of [18F]fluoro-L-DOPA PET in the diagnosis of hyperinsulinism of infancy. Endocr Dev. 2007;12:55–66.PubMed
101.
Zurück zum Zitat Barthlen W, et al. Evaluation of [18F]fluoro-L-DOPA positron emission tomography-computed tomography for surgery in focal congenital hyperinsulinism. J Clin Endocrinol Metab. 2008;93(3):869–75.PubMed Barthlen W, et al. Evaluation of [18F]fluoro-L-DOPA positron emission tomography-computed tomography for surgery in focal congenital hyperinsulinism. J Clin Endocrinol Metab. 2008;93(3):869–75.PubMed
102.
Zurück zum Zitat Zani A, et al. The predictive value of preoperative fluorine-18-L-3,4-dihydroxyphenylalanine positron emission tomography-computed tomography scans in children with congenital hyperinsulinism of infancy. J Pediatr Surg. 2011;46(1):204–8.PubMed Zani A, et al. The predictive value of preoperative fluorine-18-L-3,4-dihydroxyphenylalanine positron emission tomography-computed tomography scans in children with congenital hyperinsulinism of infancy. J Pediatr Surg. 2011;46(1):204–8.PubMed
103.
Zurück zum Zitat Capito C, Khen-Dunlop N, Ribeiro MJ, Brunelle F, Aigrain Y, Crétolle C, et al. Value of 18F-fluoro-L-dopa PET in the preoperative localization of focal lesions in congenital hyperinsulinism. Radiology. 2009;253(1):216–22.PubMed Capito C, Khen-Dunlop N, Ribeiro MJ, Brunelle F, Aigrain Y, Crétolle C, et al. Value of 18F-fluoro-L-dopa PET in the preoperative localization of focal lesions in congenital hyperinsulinism. Radiology. 2009;253(1):216–22.PubMed
104.
Zurück zum Zitat Treglia G, Mirk P, Giordano A, Rufini V. Diagnostic performance of fluorine-18-dihydroxyphenylalanine positron emission tomography in diagnosing and localizing the focal form of congenital hyperinsulinism: a meta-analysis. Pediatr Radiol. 2012;42(11):1372–9.PubMed Treglia G, Mirk P, Giordano A, Rufini V. Diagnostic performance of fluorine-18-dihydroxyphenylalanine positron emission tomography in diagnosing and localizing the focal form of congenital hyperinsulinism: a meta-analysis. Pediatr Radiol. 2012;42(11):1372–9.PubMed
105.
Zurück zum Zitat Garg PK, et al. Pancreatic uptake and radiation dosimetry of 6-[18F]fluoro-L-DOPA from PET imaging studies in infants with congenital hyperinsulinism. PLoS One. 2017;12(11):e0186340.PubMedPubMedCentral Garg PK, et al. Pancreatic uptake and radiation dosimetry of 6-[18F]fluoro-L-DOPA from PET imaging studies in infants with congenital hyperinsulinism. PLoS One. 2017;12(11):e0186340.PubMedPubMedCentral
106.
Zurück zum Zitat Maines E, Giacomello L, D'Onofrio M, Salgarello M, Gaudino R, Baggio L, et al. Images from. Nucl Med Mol Imaging. 2017;51(4):362–3.PubMed Maines E, Giacomello L, D'Onofrio M, Salgarello M, Gaudino R, Baggio L, et al. Images from. Nucl Med Mol Imaging. 2017;51(4):362–3.PubMed
107.
Zurück zum Zitat Kühnen P, Matthae R, Arya V, Hauptmann K, Rothe K, Wächter S, et al. Occurrence of giant focal forms of congenital hyperinsulinism with incorrect visualization by (18) F DOPA-PET/CT scanning. Clin Endocrinol. 2014;81(6):847–54. Kühnen P, Matthae R, Arya V, Hauptmann K, Rothe K, Wächter S, et al. Occurrence of giant focal forms of congenital hyperinsulinism with incorrect visualization by (18) F DOPA-PET/CT scanning. Clin Endocrinol. 2014;81(6):847–54.
108.
Zurück zum Zitat Parihar AS, et al. 68Ga DOTA-Exendin PET/CT for detection of Insulinoma in a patient with persistent Hyperinsulinemic hypoglycemia. Clin Nucl Med. 2018;43(8):e285–e286. Parihar AS, et al. 68Ga DOTA-Exendin PET/CT for detection of Insulinoma in a patient with persistent Hyperinsulinemic hypoglycemia. Clin Nucl Med. 2018;43(8):e285–e286.
109.
Zurück zum Zitat Cuthbertson DJ, Banks M, Khoo B, Antwi K, Christ E, Campbell F, et al. Application of Ga(68) -DOTA-exendin-4 PET/CT to localize an occult insulinoma. Clin Endocrinol. 2016;84(5):789–91. Cuthbertson DJ, Banks M, Khoo B, Antwi K, Christ E, Campbell F, et al. Application of Ga(68) -DOTA-exendin-4 PET/CT to localize an occult insulinoma. Clin Endocrinol. 2016;84(5):789–91.
110.
Zurück zum Zitat Tuzcu SA, Pekkolay Z, Kılınç F, Tuzcu AK. Ga-DOTATATE PET/CT can be an alternative imaging method in Insulinoma patients. J Nucl Med Technol. 2017;45(3):198–200.PubMed Tuzcu SA, Pekkolay Z, Kılınç F, Tuzcu AK. Ga-DOTATATE PET/CT can be an alternative imaging method in Insulinoma patients. J Nucl Med Technol. 2017;45(3):198–200.PubMed
111.
Zurück zum Zitat Deppen SA, Blume J, Bobbey AJ, Shah C, Graham MM, Lee P, et al. 68Ga-DOTATATE compared with 111In-DTPA-Octreotide and conventional imaging for pulmonary and Gastroenteropancreatic neuroendocrine tumors: a systematic review and meta-analysis. J Nucl Med. 2016;57(6):872–8.PubMedPubMedCentral Deppen SA, Blume J, Bobbey AJ, Shah C, Graham MM, Lee P, et al. 68Ga-DOTATATE compared with 111In-DTPA-Octreotide and conventional imaging for pulmonary and Gastroenteropancreatic neuroendocrine tumors: a systematic review and meta-analysis. J Nucl Med. 2016;57(6):872–8.PubMedPubMedCentral
112.
Zurück zum Zitat Sharma P, et al. Somatostatin receptor based PET/CT imaging with 68Ga-DOTA-Nal3-octreotide for localization of clinically and biochemically suspected insulinoma. Q J Nucl Med Mol Imaging. 2016;60(1):69–76.PubMed Sharma P, et al. Somatostatin receptor based PET/CT imaging with 68Ga-DOTA-Nal3-octreotide for localization of clinically and biochemically suspected insulinoma. Q J Nucl Med Mol Imaging. 2016;60(1):69–76.PubMed
113.
Zurück zum Zitat Hussain K, Blankenstein O, de Lonlay P, Christesen HT. Hyperinsulinaemic hypoglycaemia: biochemical basis and the importance of maintaining normoglycaemia during management. Arch Dis Child. 2007;92(7):568–70.PubMedPubMedCentral Hussain K, Blankenstein O, de Lonlay P, Christesen HT. Hyperinsulinaemic hypoglycaemia: biochemical basis and the importance of maintaining normoglycaemia during management. Arch Dis Child. 2007;92(7):568–70.PubMedPubMedCentral
114.
Zurück zum Zitat Nebesio TD, Hoover WC, Caldwell RL, Nitu ME, Eugster EA. Development of pulmonary hypertension in an infant treated with diazoxide. J Pediatr Endocrinol Metab. 2007;20(8):939–44. Nebesio TD, Hoover WC, Caldwell RL, Nitu ME, Eugster EA. Development of pulmonary hypertension in an infant treated with diazoxide. J Pediatr Endocrinol Metab. 2007;20(8):939–44.
115.
Zurück zum Zitat Timlin MR, Black AB, Delaney HM, Matos RI, Percival CS. Development of pulmonary hypertension during treatment with Diazoxide: a case series and literature review. Pediatr Cardiol. 2017;38(6):1247–50. Timlin MR, Black AB, Delaney HM, Matos RI, Percival CS. Development of pulmonary hypertension during treatment with Diazoxide: a case series and literature review. Pediatr Cardiol. 2017;38(6):1247–50.
116.
Zurück zum Zitat Chen SC, Dastamani A, Pintus D, Yau D, Aftab S, Bath L, et al. Diazoxide‐induced pulmonary hypertension in hyperinsulinaemic hypoglycaemia: Recommendations from a multicentre study in the United Kingdom. Clin Endocrinol. 2019;91(6):770–775. Chen SC, Dastamani A, Pintus D, Yau D, Aftab S, Bath L, et al. Diazoxide‐induced pulmonary hypertension in hyperinsulinaemic hypoglycaemia: Recommendations from a multicentre study in the United Kingdom. Clin Endocrinol. 2019;91(6):770–775.
117.
Zurück zum Zitat Arnoux JB, et al. Congenital hyperinsulinism: current trends in diagnosis and therapy. Orphanet J Rare Dis. 2011;6:63.PubMedPubMedCentral Arnoux JB, et al. Congenital hyperinsulinism: current trends in diagnosis and therapy. Orphanet J Rare Dis. 2011;6:63.PubMedPubMedCentral
118.
Zurück zum Zitat Müller D, Zimmering M, Roehr CC. Should nifedipine be used to counter low blood sugar levels in children with persistent hyperinsulinaemic hypoglycaemia? Arch Dis Child. 2004;89(1):83–5.PubMedPubMedCentral Müller D, Zimmering M, Roehr CC. Should nifedipine be used to counter low blood sugar levels in children with persistent hyperinsulinaemic hypoglycaemia? Arch Dis Child. 2004;89(1):83–5.PubMedPubMedCentral
119.
Zurück zum Zitat Baş F, et al. Successful therapy with calcium channel blocker (nifedipine) in persistent neonatal hyperinsulinemic hypoglycemia of infancy. J Pediatr Endocrinol Metab. 1999;12(6):873–8.PubMed Baş F, et al. Successful therapy with calcium channel blocker (nifedipine) in persistent neonatal hyperinsulinemic hypoglycemia of infancy. J Pediatr Endocrinol Metab. 1999;12(6):873–8.PubMed
120.
Zurück zum Zitat Shanbag P, Pathak A, Vaidya M, Shahid SK. Persistent hyperinsulinemic hypoglycemia of infancy--successful therapy with nifedipine. Indian J Pediatr. 2002;69(3):271–2.PubMed Shanbag P, Pathak A, Vaidya M, Shahid SK. Persistent hyperinsulinemic hypoglycemia of infancy--successful therapy with nifedipine. Indian J Pediatr. 2002;69(3):271–2.PubMed
121.
Zurück zum Zitat Eichmann D, Hufnagel M, Quick P, Santer R. Treatment of hyperinsulinaemic hypoglycaemia with nifedipine. Eur J Pediatr. 1999;158(3):204–6.PubMed Eichmann D, Hufnagel M, Quick P, Santer R. Treatment of hyperinsulinaemic hypoglycaemia with nifedipine. Eur J Pediatr. 1999;158(3):204–6.PubMed
122.
Zurück zum Zitat Welters A, et al. Long-term medical treatment in congenital hyperinsulinism: a descriptive analysis in a large cohort of patients from different clinical centers. Orphanet J Rare Dis. 2015;10:150.PubMedPubMedCentral Welters A, et al. Long-term medical treatment in congenital hyperinsulinism: a descriptive analysis in a large cohort of patients from different clinical centers. Orphanet J Rare Dis. 2015;10:150.PubMedPubMedCentral
123.
Zurück zum Zitat Güemes M, Shah P, Silvera S, Morgan K, Gilbert C, Hinchey L, et al. Assessment of Nifedipine therapy in Hyperinsulinemic hypoglycemia due to mutations in the ABCC8 gene. J Clin Endocrinol Metab. 2017;102(3):822–30.PubMed Güemes M, Shah P, Silvera S, Morgan K, Gilbert C, Hinchey L, et al. Assessment of Nifedipine therapy in Hyperinsulinemic hypoglycemia due to mutations in the ABCC8 gene. J Clin Endocrinol Metab. 2017;102(3):822–30.PubMed
124.
Zurück zum Zitat Durmaz E, Flanagan SE, Parlak M, Ellard S, Akcurin S, Bircan I. A combination of nifedipine and octreotide treatment in an hyperinsulinemic hypoglycemic infant. J Clin Res Pediatr Endocrinol. 2014;6(2):119–21.PubMedPubMedCentral Durmaz E, Flanagan SE, Parlak M, Ellard S, Akcurin S, Bircan I. A combination of nifedipine and octreotide treatment in an hyperinsulinemic hypoglycemic infant. J Clin Res Pediatr Endocrinol. 2014;6(2):119–21.PubMedPubMedCentral
125.
Zurück zum Zitat Glaser B, Hirsch HJ, Landau H. Persistent hyperinsulinemic hypoglycemia of infancy: long-term octreotide treatment without pancreatectomy. J Pediatr. 1993;123(4):644–50.PubMed Glaser B, Hirsch HJ, Landau H. Persistent hyperinsulinemic hypoglycemia of infancy: long-term octreotide treatment without pancreatectomy. J Pediatr. 1993;123(4):644–50.PubMed
126.
Zurück zum Zitat Thornton PS, Alter CA, Katz LE, Baker L, Stanley CA. Short- and long-term use of octreotide in the treatment of congenital hyperinsulinism. J Pediatr. 1993;123(4):637–43.PubMed Thornton PS, Alter CA, Katz LE, Baker L, Stanley CA. Short- and long-term use of octreotide in the treatment of congenital hyperinsulinism. J Pediatr. 1993;123(4):637–43.PubMed
127.
Zurück zum Zitat Roženková K, et al. The diagnosis and Management of Hyperinsulinaemic Hypoglycaemia. J Clin Res Pediatr Endocrinol. 2015;7(2):86–97.PubMedPubMedCentral Roženková K, et al. The diagnosis and Management of Hyperinsulinaemic Hypoglycaemia. J Clin Res Pediatr Endocrinol. 2015;7(2):86–97.PubMedPubMedCentral
128.
Zurück zum Zitat Mohnike K, Blankenstein O, Pfuetzner A, Pötzsch S, Schober E, Steiner S, et al. Long-term non-surgical therapy of severe persistent congenital hyperinsulinism with glucagon. Horm Res. 2008;70(1):59–64.PubMed Mohnike K, Blankenstein O, Pfuetzner A, Pötzsch S, Schober E, Steiner S, et al. Long-term non-surgical therapy of severe persistent congenital hyperinsulinism with glucagon. Horm Res. 2008;70(1):59–64.PubMed
129.
Zurück zum Zitat Brun JF, Fedou C, Mercier J. Postprandial reactive hypoglycemia. Diabetes Metab. 2000;26(5):337–51.PubMed Brun JF, Fedou C, Mercier J. Postprandial reactive hypoglycemia. Diabetes Metab. 2000;26(5):337–51.PubMed
130.
Zurück zum Zitat Salvatore T, Giugliano D. Pharmacokinetic-pharmacodynamic relationships of Acarbose. Clin Pharmacokinet. 1996;30(2):94–106.PubMed Salvatore T, Giugliano D. Pharmacokinetic-pharmacodynamic relationships of Acarbose. Clin Pharmacokinet. 1996;30(2):94–106.PubMed
131.
Zurück zum Zitat Le Quan Sang KH, et al. Successful treatment of congenital hyperinsulinism with long-acting release octreotide. Eur J Endocrinol. 2012;166(2):333–9.PubMed Le Quan Sang KH, et al. Successful treatment of congenital hyperinsulinism with long-acting release octreotide. Eur J Endocrinol. 2012;166(2):333–9.PubMed
132.
Zurück zum Zitat Modan-Moses D, Koren I, Mazor-Aronovitch K, Pinhas-Hamiel O, Landau H. Treatment of congenital hyperinsulinism with lanreotide acetate (Somatuline autogel). J Clin Endocrinol Metab. 2011;96(8):2312–7.PubMed Modan-Moses D, Koren I, Mazor-Aronovitch K, Pinhas-Hamiel O, Landau H. Treatment of congenital hyperinsulinism with lanreotide acetate (Somatuline autogel). J Clin Endocrinol Metab. 2011;96(8):2312–7.PubMed
133.
Zurück zum Zitat Shah P, Rahman SA, McElroy S, Gilbert C, Morgan K, Hinchey L, et al. Use of long-acting Somatostatin analogue (Lanreotide) in an adolescent with Diazoxide-responsive congenital Hyperinsulinism and its psychological impact. Horm Res Paediatr. 2015;84(5):355–60.PubMed Shah P, Rahman SA, McElroy S, Gilbert C, Morgan K, Hinchey L, et al. Use of long-acting Somatostatin analogue (Lanreotide) in an adolescent with Diazoxide-responsive congenital Hyperinsulinism and its psychological impact. Horm Res Paediatr. 2015;84(5):355–60.PubMed
134.
Zurück zum Zitat Kühnen P, et al. Long-term lanreotide treatment in six patients with congenital hyperinsulinism. Horm Res Paediatr. 2012;78(2):106–12.PubMed Kühnen P, et al. Long-term lanreotide treatment in six patients with congenital hyperinsulinism. Horm Res Paediatr. 2012;78(2):106–12.PubMed
135.
Zurück zum Zitat van der Steen I, van Albada M, Mohnike K, Christesen HT, Empting S, Salomon-Estebanez M, et al. A multicenter experience with long-acting Somatostatin analogues in patients with congenital Hyperinsulinism. Horm Res Paediatr. 2018;89(2):82–9.PubMed van der Steen I, van Albada M, Mohnike K, Christesen HT, Empting S, Salomon-Estebanez M, et al. A multicenter experience with long-acting Somatostatin analogues in patients with congenital Hyperinsulinism. Horm Res Paediatr. 2018;89(2):82–9.PubMed
136.
Zurück zum Zitat Corda H, et al. Treatment with long-acting lanreotide autogel in early infancy in patients with severe neonatal hyperinsulinism. Orphanet J Rare Dis. 2017;12(1):108.PubMedPubMedCentral Corda H, et al. Treatment with long-acting lanreotide autogel in early infancy in patients with severe neonatal hyperinsulinism. Orphanet J Rare Dis. 2017;12(1):108.PubMedPubMedCentral
137.
Zurück zum Zitat Dastamani A, Güemes M, Pitfield C, Morgan K, Rajab M, Rottenburger C, et al. The use of a long-acting Somatostatin analogue (Lanreotide) in three children with focal forms of congenital Hyperinsulinaemic Hypoglycaemia. Horm Res Paediatr. 2019;91(1):56–61.PubMed Dastamani A, Güemes M, Pitfield C, Morgan K, Rajab M, Rottenburger C, et al. The use of a long-acting Somatostatin analogue (Lanreotide) in three children with focal forms of congenital Hyperinsulinaemic Hypoglycaemia. Horm Res Paediatr. 2019;91(1):56–61.PubMed
138.
Zurück zum Zitat Kulke MH, Bergsland EK, Yao JC. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med. 2009;360(2):195–7.PubMed Kulke MH, Bergsland EK, Yao JC. Glycemic control in patients with insulinoma treated with everolimus. N Engl J Med. 2009;360(2):195–7.PubMed
139.
Zurück zum Zitat Alexandrescu S, et al. Persistent hyperinsulinemic hypoglycemia of infancy: constitutive activation of the mTOR pathway with associated exocrine-islet transdifferentiation and therapeutic implications. Int J Clin Exp Pathol. 2010;3(7):691–705.PubMedPubMedCentral Alexandrescu S, et al. Persistent hyperinsulinemic hypoglycemia of infancy: constitutive activation of the mTOR pathway with associated exocrine-islet transdifferentiation and therapeutic implications. Int J Clin Exp Pathol. 2010;3(7):691–705.PubMedPubMedCentral
140.
Zurück zum Zitat Senniappan S, Alexandrescu S, Tatevian N, Shah P, Arya V, Flanagan S, et al. Sirolimus therapy in infants with severe hyperinsulinemic hypoglycemia. N Engl J Med. 2014;370(12):1131–7.PubMed Senniappan S, Alexandrescu S, Tatevian N, Shah P, Arya V, Flanagan S, et al. Sirolimus therapy in infants with severe hyperinsulinemic hypoglycemia. N Engl J Med. 2014;370(12):1131–7.PubMed
141.
Zurück zum Zitat Guemes M, et al. Severe Hyperinsulinaemic Hypoglycaemia in Beckwith-Wiedemann syndrome due to paternal Uniparental Disomy of 11p15.5 managed with Sirolimus therapy. Horm Res Paediatr. 2016;85(5):353–7.PubMed Guemes M, et al. Severe Hyperinsulinaemic Hypoglycaemia in Beckwith-Wiedemann syndrome due to paternal Uniparental Disomy of 11p15.5 managed with Sirolimus therapy. Horm Res Paediatr. 2016;85(5):353–7.PubMed
142.
Zurück zum Zitat Shah P, Arya VB, Flanagan SE, Morgan K, Ellard S, Senniappan S, et al. Sirolimus therapy in a patient with severe hyperinsulinaemic hypoglycaemia due to a compound heterozygous ABCC8 gene mutation. J Pediatr Endocrinol Metab. 2015;28(5–6):695–9.PubMed Shah P, Arya VB, Flanagan SE, Morgan K, Ellard S, Senniappan S, et al. Sirolimus therapy in a patient with severe hyperinsulinaemic hypoglycaemia due to a compound heterozygous ABCC8 gene mutation. J Pediatr Endocrinol Metab. 2015;28(5–6):695–9.PubMed
143.
Zurück zum Zitat Al-Balwi R, et al. Sirolimus in the treatment of three infants with diffuse congenital hyperinsulinism. J Pediatr Endocrinol Metab. 2017;30(9):1013–7.PubMed Al-Balwi R, et al. Sirolimus in the treatment of three infants with diffuse congenital hyperinsulinism. J Pediatr Endocrinol Metab. 2017;30(9):1013–7.PubMed
144.
Zurück zum Zitat Minute M, et al. Sirolimus therapy in congenital Hyperinsulinism: a successful experience beyond infancy. Pediatrics. 2015;136(5):e1373–6.PubMed Minute M, et al. Sirolimus therapy in congenital Hyperinsulinism: a successful experience beyond infancy. Pediatrics. 2015;136(5):e1373–6.PubMed
145.
Zurück zum Zitat Méder Ü, et al. Severe Hyperinsulinemic hypoglycemia in a neonate: response to Sirolimus therapy. Pediatrics. 2015;136(5):e1369–72.PubMed Méder Ü, et al. Severe Hyperinsulinemic hypoglycemia in a neonate: response to Sirolimus therapy. Pediatrics. 2015;136(5):e1369–72.PubMed
146.
Zurück zum Zitat Haliloğlu B, Tüzün H, Flanagan SE, et al. Sirolimus-Induced hepatitis in two patients with hyperinsulinemic hypoglycemia. J Clin Res Pediatr Endocrinol. 2018;10(3):279–283. Haliloğlu B, Tüzün H, Flanagan SE, et al. Sirolimus-Induced hepatitis in two patients with hyperinsulinemic hypoglycemia. J Clin Res Pediatr Endocrinol. 2018;10(3):279–283.
147.
Zurück zum Zitat Dastamani A, Güemes M, Walker J, Shah P, Hussain K. Sirolimus precipitating diabetes mellitus in a patient with congenital hyperinsulinaemic hypoglycaemia due to autosomal dominant ABCC8 mutation. J Pediatr Endocrinol Metab. 2017;30(11):1219–22.PubMed Dastamani A, Güemes M, Walker J, Shah P, Hussain K. Sirolimus precipitating diabetes mellitus in a patient with congenital hyperinsulinaemic hypoglycaemia due to autosomal dominant ABCC8 mutation. J Pediatr Endocrinol Metab. 2017;30(11):1219–22.PubMed
148.
Zurück zum Zitat Szymanowski M, Estebanez MS, Padidela R, Han B, Mosinska K, Stevens A, et al. mTOR inhibitors for the treatment of severe congenital Hyperinsulinism: perspectives on limited therapeutic success. J Clin Endocrinol Metab. 2016;101(12):4719–29.PubMed Szymanowski M, Estebanez MS, Padidela R, Han B, Mosinska K, Stevens A, et al. mTOR inhibitors for the treatment of severe congenital Hyperinsulinism: perspectives on limited therapeutic success. J Clin Endocrinol Metab. 2016;101(12):4719–29.PubMed
149.
Zurück zum Zitat Banerjee I, De Leon D, Dunne MJ. Extreme caution on the use of sirolimus for the congenital hyperinsulinism in infancy patient. Orphanet J Rare Dis. 2017;12(1):70.PubMedPubMedCentral Banerjee I, De Leon D, Dunne MJ. Extreme caution on the use of sirolimus for the congenital hyperinsulinism in infancy patient. Orphanet J Rare Dis. 2017;12(1):70.PubMedPubMedCentral
150.
Zurück zum Zitat Maria G, Antonia D, Michael A, Kate M, Sian E, Sarah FE, et al. Sirolimus: efficacy and complications in children with Hyperinsulinemic hypoglycemia: a 5-year follow-up study. J Endocr Soc. 2019;3(4):699–713.PubMedPubMedCentral Maria G, Antonia D, Michael A, Kate M, Sian E, Sarah FE, et al. Sirolimus: efficacy and complications in children with Hyperinsulinemic hypoglycemia: a 5-year follow-up study. J Endocr Soc. 2019;3(4):699–713.PubMedPubMedCentral
151.
Zurück zum Zitat McClenaghan NH, Flatt PR, Ball AJ. Actions of glucagon-like peptide-1 on KATP channel-dependent and -independent effects of glucose, sulphonylureas and nateglinide. J Endocrinol. 2006;190(3):889–96.PubMed McClenaghan NH, Flatt PR, Ball AJ. Actions of glucagon-like peptide-1 on KATP channel-dependent and -independent effects of glucose, sulphonylureas and nateglinide. J Endocrinol. 2006;190(3):889–96.PubMed
152.
Zurück zum Zitat De León DD, et al. Exendin-(9-39) corrects fasting hypoglycemia in SUR-1−/− mice by lowering cAMP in pancreatic beta-cells and inhibiting insulin secretion. J Biol Chem. 2008;283(38):25786–93.PubMedPubMedCentral De León DD, et al. Exendin-(9-39) corrects fasting hypoglycemia in SUR-1−/− mice by lowering cAMP in pancreatic beta-cells and inhibiting insulin secretion. J Biol Chem. 2008;283(38):25786–93.PubMedPubMedCentral
153.
Zurück zum Zitat Calabria AC, Li C, Gallagher PR, Stanley CA, de León DD. GLP-1 receptor antagonist exendin-(9-39) elevates fasting blood glucose levels in congenital hyperinsulinism owing to inactivating mutations in the ATP-sensitive K+ channel. Diabetes. 2012;61(10):2585–91.PubMedPubMedCentral Calabria AC, Li C, Gallagher PR, Stanley CA, de León DD. GLP-1 receptor antagonist exendin-(9-39) elevates fasting blood glucose levels in congenital hyperinsulinism owing to inactivating mutations in the ATP-sensitive K+ channel. Diabetes. 2012;61(10):2585–91.PubMedPubMedCentral
154.
Zurück zum Zitat Chen PC, Olson EM, Zhou Q, Kryukova Y, Sampson HM, Thomas DY, et al. Carbamazepine as a novel small molecule corrector of trafficking-impaired ATP-sensitive potassium channels identified in congenital hyperinsulinism. J Biol Chem. 2013;288(29):20942–54.PubMedPubMedCentral Chen PC, Olson EM, Zhou Q, Kryukova Y, Sampson HM, Thomas DY, et al. Carbamazepine as a novel small molecule corrector of trafficking-impaired ATP-sensitive potassium channels identified in congenital hyperinsulinism. J Biol Chem. 2013;288(29):20942–54.PubMedPubMedCentral
155.
Zurück zum Zitat Yan F, Lin CW, Weisiger E, Cartier EA, Taschenberger G, Shyng SL. Sulfonylureas correct trafficking defects of ATP-sensitive potassium channels caused by mutations in the sulfonylurea receptor. J Biol Chem. 2004;279(12):11096–105.PubMed Yan F, Lin CW, Weisiger E, Cartier EA, Taschenberger G, Shyng SL. Sulfonylureas correct trafficking defects of ATP-sensitive potassium channels caused by mutations in the sulfonylurea receptor. J Biol Chem. 2004;279(12):11096–105.PubMed
156.
Zurück zum Zitat Devaraneni PK, Martin GM, Olson EM, Zhou Q, Shyng SL. Structurally distinct ligands rescue biogenesis defects of the KATP channel complex via a converging mechanism. J Biol Chem. 2015;290(12):7980–91.PubMedPubMedCentral Devaraneni PK, Martin GM, Olson EM, Zhou Q, Shyng SL. Structurally distinct ligands rescue biogenesis defects of the KATP channel complex via a converging mechanism. J Biol Chem. 2015;290(12):7980–91.PubMedPubMedCentral
157.
Zurück zum Zitat Martin GM, Rex EA, Devaraneni P, Denton JS, Boodhansingh KE, DeLeon D, et al. Pharmacological correction of trafficking defects in ATP-sensitive potassium channels caused by sulfonylurea receptor 1 mutations. J Biol Chem. 2016;291(42):21971–83.PubMedPubMedCentral Martin GM, Rex EA, Devaraneni P, Denton JS, Boodhansingh KE, DeLeon D, et al. Pharmacological correction of trafficking defects in ATP-sensitive potassium channels caused by sulfonylurea receptor 1 mutations. J Biol Chem. 2016;291(42):21971–83.PubMedPubMedCentral
158.
Zurück zum Zitat Neylon OM, Moran MM, Pellicano A, Nightingale M, O'Connell MA. Successful subcutaneous glucagon use for persistent hypoglycaemia in congenital hyperinsulinism. J Pediatr Endocrinol Metab. 2013;26(11–12):1157–61.PubMed Neylon OM, Moran MM, Pellicano A, Nightingale M, O'Connell MA. Successful subcutaneous glucagon use for persistent hypoglycaemia in congenital hyperinsulinism. J Pediatr Endocrinol Metab. 2013;26(11–12):1157–61.PubMed
159.
Zurück zum Zitat Hövelmann U, Bysted BV, Mouritzen U, Macchi F, Lamers D, Kronshage B, et al. Pharmacokinetic and Pharmacodynamic characteristics of Dasiglucagon, a novel soluble and stable glucagon analog. Diabetes Care. 2018;41(3):531–7.PubMed Hövelmann U, Bysted BV, Mouritzen U, Macchi F, Lamers D, Kronshage B, et al. Pharmacokinetic and Pharmacodynamic characteristics of Dasiglucagon, a novel soluble and stable glucagon analog. Diabetes Care. 2018;41(3):531–7.PubMed
160.
Zurück zum Zitat Laguna Sanz AJ, Mulla CM, Fowler KM, Cloutier E, Goldfine AB, Newswanger B, et al. Design and clinical evaluation of a novel low-glucose prediction algorithm with mini-dose stable glucagon delivery in post-bariatric hypoglycemia. Diabetes Technol Ther. 2018;20(2):127–39.PubMedPubMedCentral Laguna Sanz AJ, Mulla CM, Fowler KM, Cloutier E, Goldfine AB, Newswanger B, et al. Design and clinical evaluation of a novel low-glucose prediction algorithm with mini-dose stable glucagon delivery in post-bariatric hypoglycemia. Diabetes Technol Ther. 2018;20(2):127–39.PubMedPubMedCentral
161.
Zurück zum Zitat Pierro A, Nah SA. Surgical management of congenital hyperinsulinism of infancy. Semin Pediatr Surg. 2011;20(1):50–3.PubMed Pierro A, Nah SA. Surgical management of congenital hyperinsulinism of infancy. Semin Pediatr Surg. 2011;20(1):50–3.PubMed
162.
Zurück zum Zitat Adzick NS, Thornton PS, Stanley CA, Kaye RD, Ruchelli E. A multidisciplinary approach to the focal form of congenital hyperinsulinism leads to successful treatment by partial pancreatectomy. J Pediatr Surg. 2004;39(3):270–5.PubMed Adzick NS, Thornton PS, Stanley CA, Kaye RD, Ruchelli E. A multidisciplinary approach to the focal form of congenital hyperinsulinism leads to successful treatment by partial pancreatectomy. J Pediatr Surg. 2004;39(3):270–5.PubMed
163.
Zurück zum Zitat Al-Shanafey S. Laparoscopic vs open pancreatectomy for persistent hyperinsulinemic hypoglycemia of infancy. J Pediatr Surg. 2009;44(5):957–61.PubMed Al-Shanafey S. Laparoscopic vs open pancreatectomy for persistent hyperinsulinemic hypoglycemia of infancy. J Pediatr Surg. 2009;44(5):957–61.PubMed
164.
Zurück zum Zitat Laje P, et al. Pancreatic head resection and roux-en-Y pancreaticojejunostomy for the treatment of the focal form of congenital hyperinsulinism. J Pediatr Surg. 2012;47(1):130–5.PubMedPubMedCentral Laje P, et al. Pancreatic head resection and roux-en-Y pancreaticojejunostomy for the treatment of the focal form of congenital hyperinsulinism. J Pediatr Surg. 2012;47(1):130–5.PubMedPubMedCentral
165.
Zurück zum Zitat Beltrand J, Caquard M, Arnoux JB, Laborde K, Velho G, Verkarre V, et al. Glucose metabolism in 105 children and adolescents after pancreatectomy for congenital hyperinsulinism. Diabetes Care. 2012;35(2):198–203.PubMedPubMedCentral Beltrand J, Caquard M, Arnoux JB, Laborde K, Velho G, Verkarre V, et al. Glucose metabolism in 105 children and adolescents after pancreatectomy for congenital hyperinsulinism. Diabetes Care. 2012;35(2):198–203.PubMedPubMedCentral
166.
Zurück zum Zitat Arya VB, Senniappan S, Demirbilek H, Alam S, Flanagan SE, Ellard S, et al. Pancreatic endocrine and exocrine function in children following near-total pancreatectomy for diffuse congenital hyperinsulinism. PLoS One. 2014;9(5):e98054.PubMedPubMedCentral Arya VB, Senniappan S, Demirbilek H, Alam S, Flanagan SE, Ellard S, et al. Pancreatic endocrine and exocrine function in children following near-total pancreatectomy for diffuse congenital hyperinsulinism. PLoS One. 2014;9(5):e98054.PubMedPubMedCentral
167.
Zurück zum Zitat Shah P, Demirbilek H, Hussain K. Persistent hyperinsulinaemic hypoglycaemia in infancy. Semin Pediatr Surg. 2014;23(2):76–82.PubMed Shah P, Demirbilek H, Hussain K. Persistent hyperinsulinaemic hypoglycaemia in infancy. Semin Pediatr Surg. 2014;23(2):76–82.PubMed
168.
Zurück zum Zitat Gouya H, Vignaux O, Augui J, Dousset B, Palazzo L, Louvel A, et al. CT, endoscopic sonography, and a combined protocol for preoperative evaluation of pancreatic insulinomas. AJR Am J Roentgenol. 2003;181(4):987–92.PubMed Gouya H, Vignaux O, Augui J, Dousset B, Palazzo L, Louvel A, et al. CT, endoscopic sonography, and a combined protocol for preoperative evaluation of pancreatic insulinomas. AJR Am J Roentgenol. 2003;181(4):987–92.PubMed
169.
Zurück zum Zitat Iglesias P, Lafuente C, Martín Almendra MÁ, López Guzmán A, Castro JC, Díez JJ. Insulinoma: a multicenter, retrospective analysis of three decades of experience (1983-2014). Endocrinol Nutr. 2015;62(7):306–13.PubMed Iglesias P, Lafuente C, Martín Almendra MÁ, López Guzmán A, Castro JC, Díez JJ. Insulinoma: a multicenter, retrospective analysis of three decades of experience (1983-2014). Endocrinol Nutr. 2015;62(7):306–13.PubMed
170.
Zurück zum Zitat Okabayashi T, Shima Y, Sumiyoshi T, Kozuki A, Ito S, Ogawa Y, et al. Diagnosis and management of insulinoma. World J Gastroenterol. 2013;19(6):829–37.PubMedPubMedCentral Okabayashi T, Shima Y, Sumiyoshi T, Kozuki A, Ito S, Ogawa Y, et al. Diagnosis and management of insulinoma. World J Gastroenterol. 2013;19(6):829–37.PubMedPubMedCentral
171.
Zurück zum Zitat Antonakis PT, Ashrafian H, Martinez-Isla A. Pancreatic insulinomas: laparoscopic management. World J Gastrointest Endosc. 2015;7(16):1197–207.PubMedPubMedCentral Antonakis PT, Ashrafian H, Martinez-Isla A. Pancreatic insulinomas: laparoscopic management. World J Gastrointest Endosc. 2015;7(16):1197–207.PubMedPubMedCentral
172.
Zurück zum Zitat Ito T, Igarashi H, Jensen RT. Pancreatic neuroendocrine tumors: clinical features, diagnosis and medical treatment: advances. Best Pract Res Clin Gastroenterol. 2012;26(6):737–53.PubMedPubMedCentral Ito T, Igarashi H, Jensen RT. Pancreatic neuroendocrine tumors: clinical features, diagnosis and medical treatment: advances. Best Pract Res Clin Gastroenterol. 2012;26(6):737–53.PubMedPubMedCentral
173.
Zurück zum Zitat Salomon-Estebanez M, Flanagan SE, Ellard S, Rigby L, Bowden L, Mohamed Z, et al. Conservatively treated congenital Hyperinsulinism (CHI) due to K-ATP channel gene mutations: reducing severity over time. Orphanet J Rare Dis. 2016;11(1):163.PubMedPubMedCentral Salomon-Estebanez M, Flanagan SE, Ellard S, Rigby L, Bowden L, Mohamed Z, et al. Conservatively treated congenital Hyperinsulinism (CHI) due to K-ATP channel gene mutations: reducing severity over time. Orphanet J Rare Dis. 2016;11(1):163.PubMedPubMedCentral
174.
Zurück zum Zitat Sun L, Coy DH. Somatostatin and its analogs. Curr Drug Targets. 2016;17(9):529–537. Sun L, Coy DH. Somatostatin and its analogs. Curr Drug Targets. 2016;17(9):529–537.
175.
Zurück zum Zitat Demirbilek H, Shah P, Arya VB, Hinchey L, Flanagan SE, Ellard S, et al. Long-term follow-up of children with congenital hyperinsulinism on octreotide therapy. J Clin Endocrinol Metab. 2014;99(10):3660–7.PubMed Demirbilek H, Shah P, Arya VB, Hinchey L, Flanagan SE, Ellard S, et al. Long-term follow-up of children with congenital hyperinsulinism on octreotide therapy. J Clin Endocrinol Metab. 2014;99(10):3660–7.PubMed
176.
Zurück zum Zitat Lord K, et al. High risk of diabetes and neurobehavioral deficits in individuals with surgically treated Hyperinsulinism. J Clin Endocrinol Metab. 2015;100(11):4133–9.PubMedPubMedCentral Lord K, et al. High risk of diabetes and neurobehavioral deficits in individuals with surgically treated Hyperinsulinism. J Clin Endocrinol Metab. 2015;100(11):4133–9.PubMedPubMedCentral
177.
Zurück zum Zitat Ludwig A, et al. Formal neurocognitive testing in 60 patients with congenital Hyperinsulinism. Horm Res Paediatr. 2018;89(1):1–6.PubMed Ludwig A, et al. Formal neurocognitive testing in 60 patients with congenital Hyperinsulinism. Horm Res Paediatr. 2018;89(1):1–6.PubMed
179.
Zurück zum Zitat Lotstein DS, et al. Transition from pediatric to adult care for youth diagnosed with type 1 diabetes in adolescence. Pediatrics. 2013;131(4):e1062–70.PubMedPubMedCentral Lotstein DS, et al. Transition from pediatric to adult care for youth diagnosed with type 1 diabetes in adolescence. Pediatrics. 2013;131(4):e1062–70.PubMedPubMedCentral
180.
Zurück zum Zitat Viner R. Transition from paediatric to adult care. Bridging the gaps or passing the buck? Arch Dis Child. 1999;81(3):271–5.PubMedPubMedCentral Viner R. Transition from paediatric to adult care. Bridging the gaps or passing the buck? Arch Dis Child. 1999;81(3):271–5.PubMedPubMedCentral
181.
Zurück zum Zitat Zhou H, Roberts P, Dhaliwal S, Della P. Transitioning adolescent and young adults with chronic disease and/or disabilities from paediatric to adult care services - an integrative review. J Clin Nurs. 2016;25(21–22):3113–30.PubMedPubMedCentral Zhou H, Roberts P, Dhaliwal S, Della P. Transitioning adolescent and young adults with chronic disease and/or disabilities from paediatric to adult care services - an integrative review. J Clin Nurs. 2016;25(21–22):3113–30.PubMedPubMedCentral
182.
Zurück zum Zitat de Silva PS, Fishman LN. Transition of the patient with IBD from pediatric to adult care-an assessment of current evidence. Inflamm Bowel Dis. 2014;20(8):1458–64.PubMed de Silva PS, Fishman LN. Transition of the patient with IBD from pediatric to adult care-an assessment of current evidence. Inflamm Bowel Dis. 2014;20(8):1458–64.PubMed
183.
Zurück zum Zitat Borus JS, Laffel L. Adherence challenges in the management of type 1 diabetes in adolescents: prevention and intervention. Curr Opin Pediatr. 2010;22(4):405–11.PubMedPubMedCentral Borus JS, Laffel L. Adherence challenges in the management of type 1 diabetes in adolescents: prevention and intervention. Curr Opin Pediatr. 2010;22(4):405–11.PubMedPubMedCentral
184.
Zurück zum Zitat Valenzuela JM, Buchanan CL, Radcliffe J, Ambrose C, Hawkins LA, Tanney M, et al. Transition to adult services among behaviorally infected adolescents with HIV--a qualitative study. J Pediatr Psychol. 2011;36(2):134–40.PubMed Valenzuela JM, Buchanan CL, Radcliffe J, Ambrose C, Hawkins LA, Tanney M, et al. Transition to adult services among behaviorally infected adolescents with HIV--a qualitative study. J Pediatr Psychol. 2011;36(2):134–40.PubMed
185.
Zurück zum Zitat Hanna KM, Woodward J. The transition from pediatric to adult diabetes care services. Clin Nurse Spec. 2013;27(3):132–45.PubMedPubMedCentral Hanna KM, Woodward J. The transition from pediatric to adult diabetes care services. Clin Nurse Spec. 2013;27(3):132–45.PubMedPubMedCentral
186.
Zurück zum Zitat van Staa A, Sattoe JN. Young adults' experiences and satisfaction with the transfer of care. J Adolesc Health. 2014;55(6):796–803.PubMed van Staa A, Sattoe JN. Young adults' experiences and satisfaction with the transfer of care. J Adolesc Health. 2014;55(6):796–803.PubMed
187.
Zurück zum Zitat Barlow J, et al. Self-management approaches for people with chronic conditions: a review. Patient Educ Couns. 2002;48(2):177–87.PubMed Barlow J, et al. Self-management approaches for people with chronic conditions: a review. Patient Educ Couns. 2002;48(2):177–87.PubMed
188.
Zurück zum Zitat Acuña Mora M, Sparud-Lundin C, Bratt EL, Moons P. Person-centred transition programme to empower adolescents with congenital heart disease in the transition to adulthood: a study protocol for a hybrid randomised controlled trial (STEPSTONES project). BMJ Open. 2017;7(4):e014593.PubMedPubMedCentral Acuña Mora M, Sparud-Lundin C, Bratt EL, Moons P. Person-centred transition programme to empower adolescents with congenital heart disease in the transition to adulthood: a study protocol for a hybrid randomised controlled trial (STEPSTONES project). BMJ Open. 2017;7(4):e014593.PubMedPubMedCentral
189.
190.
Zurück zum Zitat Peters A, Laffel L, A.D.A.T.W. Group. Diabetes care for emerging adults: recommendations for transition from pediatric to adult diabetes care systems: a position statement of the American Diabetes Association, with representation by the American College of Osteopathic Family Physicians, the American Academy of Pediatrics, the American Association of Clinical Endocrinologists, the American Osteopathic Association, the Centers for Disease Control and Prevention, Children with Diabetes, The Endocrine Society, the International Society for Pediatric and Adolescent Diabetes, Juvenile Diabetes Research Foundation International, the National Diabetes Education Program, and the Pediatric Endocrine Society (formerly Lawson Wilkins Pediatric Endocrine Society). Diabetes Care. 2011;34(11):2477–85.PubMedPubMedCentral Peters A, Laffel L, A.D.A.T.W. Group. Diabetes care for emerging adults: recommendations for transition from pediatric to adult diabetes care systems: a position statement of the American Diabetes Association, with representation by the American College of Osteopathic Family Physicians, the American Academy of Pediatrics, the American Association of Clinical Endocrinologists, the American Osteopathic Association, the Centers for Disease Control and Prevention, Children with Diabetes, The Endocrine Society, the International Society for Pediatric and Adolescent Diabetes, Juvenile Diabetes Research Foundation International, the National Diabetes Education Program, and the Pediatric Endocrine Society (formerly Lawson Wilkins Pediatric Endocrine Society). Diabetes Care. 2011;34(11):2477–85.PubMedPubMedCentral
191.
Zurück zum Zitat Choudhary P, Amiel SA. Hypoglycaemia in type 1 diabetes: technological treatments, their limitations and the place of psychology. Diabetologia. 2018;61(4):761–9.PubMedPubMedCentral Choudhary P, Amiel SA. Hypoglycaemia in type 1 diabetes: technological treatments, their limitations and the place of psychology. Diabetologia. 2018;61(4):761–9.PubMedPubMedCentral
192.
Zurück zum Zitat Clarke WL, Cox DJ, Gonder-Frederick LA, Julian D, Schlundt D, Polonsky W. Reduced awareness of hypoglycemia in adults with IDDM. A prospective study of hypoglycemic frequency and associated symptoms. Diabetes Care. 1995;18(4):517–22.PubMed Clarke WL, Cox DJ, Gonder-Frederick LA, Julian D, Schlundt D, Polonsky W. Reduced awareness of hypoglycemia in adults with IDDM. A prospective study of hypoglycemic frequency and associated symptoms. Diabetes Care. 1995;18(4):517–22.PubMed
193.
Zurück zum Zitat Gold AE, MacLeod KM, Frier BM. Frequency of severe hypoglycemia in patients with type I diabetes with impaired awareness of hypoglycemia. Diabetes Care. 1994;17(7):697–703.PubMed Gold AE, MacLeod KM, Frier BM. Frequency of severe hypoglycemia in patients with type I diabetes with impaired awareness of hypoglycemia. Diabetes Care. 1994;17(7):697–703.PubMed
194.
Zurück zum Zitat Hay WW, et al. Knowledge gaps and research needs for understanding and treating neonatal hypoglycemia: workshop report from Eunice Kennedy Shriver National Institute of Child Health and Human Development. J Pediatr. 2009;155(5):612–7.PubMed Hay WW, et al. Knowledge gaps and research needs for understanding and treating neonatal hypoglycemia: workshop report from Eunice Kennedy Shriver National Institute of Child Health and Human Development. J Pediatr. 2009;155(5):612–7.PubMed
195.
Zurück zum Zitat DVLA Assessing fitness to drive – a guide for medical. professionals [Internet]. May 20, 2018. DVLA Assessing fitness to drive – a guide for medical. professionals [Internet]. May 20, 2018.
197.
Zurück zum Zitat Rosenbaum P, Stewart D. Perspectives on transitions: rethinking services for children and youth with developmental disabilities. Arch Phys Med Rehabil. 2007;88(8):1080–2.PubMed Rosenbaum P, Stewart D. Perspectives on transitions: rethinking services for children and youth with developmental disabilities. Arch Phys Med Rehabil. 2007;88(8):1080–2.PubMed
198.
Zurück zum Zitat Cannarella R, Arato I, Condorelli RA, et al. Effects of insulin on porcine neonatal sertoli cell responsiveness to FSH In Vitro. J Clin Med. 2019;8(6):809. Cannarella R, Arato I, Condorelli RA, et al. Effects of insulin on porcine neonatal sertoli cell responsiveness to FSH In Vitro. J Clin Med. 2019;8(6):809.
199.
Zurück zum Zitat Garvey KC, Wolpert HA, Laffel LM, Rhodes ET, Wolfsdorf JI, Finkelstein JA. Health care transition in young adults with type 1 diabetes: barriers to timely establishment of adult diabetes care. Endocr Pract. 2013;19(6):946–52.PubMedPubMedCentral Garvey KC, Wolpert HA, Laffel LM, Rhodes ET, Wolfsdorf JI, Finkelstein JA. Health care transition in young adults with type 1 diabetes: barriers to timely establishment of adult diabetes care. Endocr Pract. 2013;19(6):946–52.PubMedPubMedCentral
200.
Zurück zum Zitat Johnson SR, Leo PJ, McInerney-Leo A, Anderson LK, Marshall M, McGown I, et al. Whole-exome sequencing for mutation detection in pediatric disorders of insulin secretion: maturity onset diabetes of the young and congenital hyperinsulinism. Pediatr Diabetes. 2018;19(4):656–62.PubMed Johnson SR, Leo PJ, McInerney-Leo A, Anderson LK, Marshall M, McGown I, et al. Whole-exome sequencing for mutation detection in pediatric disorders of insulin secretion: maturity onset diabetes of the young and congenital hyperinsulinism. Pediatr Diabetes. 2018;19(4):656–62.PubMed
201.
Zurück zum Zitat Shah P, et al. Hyperinsulinaemic hypoglycaemia in children and adults. Lancet Diabetes Endocrinol. 2017;5(9):729–42.PubMed Shah P, et al. Hyperinsulinaemic hypoglycaemia in children and adults. Lancet Diabetes Endocrinol. 2017;5(9):729–42.PubMed
202.
Zurück zum Zitat Corbin JA, Bhaskar V, Goldfine ID, Issafras H, Bedinger DH, Lau A, et al. Inhibition of insulin receptor function by a human, allosteric monoclonal antibody: a potential new approach for the treatment of hyperinsulinemic hypoglycemia. MAbs. 2014;6(1):262–72.PubMed Corbin JA, Bhaskar V, Goldfine ID, Issafras H, Bedinger DH, Lau A, et al. Inhibition of insulin receptor function by a human, allosteric monoclonal antibody: a potential new approach for the treatment of hyperinsulinemic hypoglycemia. MAbs. 2014;6(1):262–72.PubMed
203.
Zurück zum Zitat Kapoor RR, James C, Hussain K. Hyperinsulinism in developmental syndromes. Endocr Dev. 2009;14:95–113.PubMed Kapoor RR, James C, Hussain K. Hyperinsulinism in developmental syndromes. Endocr Dev. 2009;14:95–113.PubMed
Metadaten
Titel
Hyperinsulinemic hypoglycemia in children and adolescents: Recent advances in understanding of pathophysiology and management
verfasst von
Maria Gϋemes
Sofia Asim Rahman
Ritika R. Kapoor
Sarah Flanagan
Jayne A. L. Houghton
Shivani Misra
Nick Oliver
Mehul Tulsidas Dattani
Pratik Shah
Publikationsdatum
17.03.2020
Verlag
Springer US
Erschienen in
Reviews in Endocrine and Metabolic Disorders / Ausgabe 4/2020
Print ISSN: 1389-9155
Elektronische ISSN: 1573-2606
DOI
https://doi.org/10.1007/s11154-020-09548-7

Weitere Artikel der Ausgabe 4/2020

Reviews in Endocrine and Metabolic Disorders 4/2020 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.