Skip to main content
Erschienen in: Diabetologia 5/2017

Open Access 17.03.2017 | Review

Precision diabetes: learning from monogenic diabetes

verfasst von: Andrew T. Hattersley, Kashyap A. Patel

Erschienen in: Diabetologia | Ausgabe 5/2017

Abstract

The precision medicine approach of tailoring treatment to the individual characteristics of each patient or subgroup has been a great success in monogenic diabetes subtypes, MODY and neonatal diabetes. This review examines what has led to the success of a precision medicine approach in monogenic diabetes (precision diabetes) and outlines possible implications for type 2 diabetes. For monogenic diabetes, the molecular genetics can define discrete aetiological subtypes that have profound implications on diabetes treatment and can predict future development of associated clinical features, allowing early preventative or supportive treatment. In contrast, type 2 diabetes has overlapping polygenic susceptibility and underlying aetiologies, making it difficult to define discrete clinical subtypes with a dramatic implication for treatment. The implementation of precision medicine in neonatal diabetes was simple and rapid as it was based on single clinical criteria (diagnosed <6 months of age). In contrast, in MODY it was more complex and slow because of the lack of single criteria to identify patients, but it was greatly assisted by the development of a diagnostic probability calculator and associated smartphone app. Experience in monogenic diabetes suggests that successful adoption of a precision diabetes approach in type 2 diabetes will require simple, quick, easily accessible stratification that is based on a combination of routine clinical data, rather than relying on newer technologies. Analysing existing clinical data from routine clinical practice and trials may provide early success for precision medicine in type 2 diabetes.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00125-017-4226-2) contains a slideset of the figures for download, which is available to authorised users.
Abkürzungen
GLP-1
Glucagon-like peptide-1
IPEX
Immunodysregulation polyendocrinopathy enteropathy X-linked
PNDM
Permanent neonatal diabetes
TNDM
Transient neonatal diabetes

Introduction

Precision medicine refers to the tailoring of medical treatment to the individual characteristics of each patient or subpopulation [1]. Precision diabetes is when a precision medicine approach is used to improve treatment of patients with diabetes. This review aims to examine how precision diabetes has been successfully applied in monogenic diabetes and to ask what this can teach us about the challenges of implementing a precision diabetes approach in type 2 diabetes. For a detailed discussion of precision diabetes for type 2 diabetes, please see review by Mark McCarthy in this issue of Diabetologia [2].

Overview of the precision medicine approach in monogenic diabetes

Overview of neonatal diabetes

Genetic causes of neonatal diabetes
Neonatal diabetes is defined as diabetes developed before 6 months of age. The knowledge that neonatal diabetes has a monogenic aetiology is based on two strong strands of evidence; first, patients with permanent diabetes diagnosed <6 months of age do not have an increased type 1 diabetes genetic susceptibility. This is in contrast with the high susceptibility seen when those diagnosed >6 months [3, 4]. Second, 96% of patients with known ‘neonatal’ monogenic genetic aetiology are diagnosed with diabetes < 6 months [5, 6]. Before genetic definition was possible, neonatal diabetes was classified solely on the clinical course of disease as transient neonatal diabetes (TNDM), permanent neonatal diabetes (PNDM), or by the specific syndrome when associated with other features e.g. Wolcott–Rallison syndrome or immunodysregulation polyendocrinopathy enteropathy X-linked (IPEX) syndrome. We now know of 23 different genetic causes of neonatal diabetes [7, 8].
Genetic diagnosis: impact on diabetes treatment
The key drive for precision diabetes has been the finding that the genetic aetiology strongly influences treatment choice and the clinical course (Fig. 1) [7]. Approximately 50% of patients with neonatal diabetes have mutations in the genes encoding the potassium channel (KCNJ11, ABCC8). These patients show excellent glucose control with high-dose sulfonylureas without an increase in hypoglycaemia and glucose variability [9]. They also show improvements in their neurological function following sulfonylurea therapy [10, 11]. Patients with transient neonatal diabetes as a result of 6q24 methylation abnormalities can be treated with low-dose sulfonylureas when they relapse (Fig. 1). In contrast, patients with other neonatal diabetes subtypes require insulin treatment.
Genetic diagnosis: impact on clinical course
Another key benefit of precision medicine is the ability to explain additional clinical abnormalities that are associated with the underlying genetic cause (Fig. 1). These may be already present (e.g. cardiac defects in patients with GATA6 mutations, microencephaly in patients with IER3IP1 mutations and gall bladder and gut atresia in patients with RFX6 mutations), anticipated (e.g. exocrine pancreas deficiency in patients with mutations in GATA4, GATA6 or PDX1, and remission of transient diabetes in patients with 6q24 methylation abnormalities) or they may develop later (e.g. hepatic failure and bone abnormalities in Wolcott–Rallison syndrome or other autoimmune conditions with IPEX syndrome) [7].
Early comprehensive genetic testing: a paradigm shift for managing neonatal diabetes
The development in targeted next-generation DNA sequencing has allowed rapid and comprehensive testing of all known genetic aetiology in monogenic diabetes [12]. In parallel, referral time from development of diabetes to genetic testing reduced from 4 years to 7 weeks between 2004 and 2013 [7]. These two factors have led to a paradigm shift in the way that we manage neonatal diabetes as we can now make a rapid and precise genetic diagnosis before the development of all clinical features (Fig. 1). This can lead to early appropriate treatment of the diabetes and future planning for other clinical developments [7]. For example, early diagnosis of TNDM allows remission to be predicted and planned for, and knowing that developmental delay is a feature of the genetic aetiology allows early developmental assessment and support. Furthermore, early treatment with sulfonylureas in KCNJ11- and ABCC8-neonatal diabetes probably results in less severe developmental delay [11]. In addition, early treatment with thiamine in thiamine-responsive megaloblastic anaemia (TRMA) neonatal diabetes can improve glycaemic control [13]. Finally, for IPEX and other severe monogenic autoimmune syndromes, early diagnosis allows consideration for early curative bone marrow transplantation before patients are too sick [14].

Overview of MODY

Genetic causes of MODY
MODY was originally defined as a clinical subgroup of familial diabetes that was diagnosed early (typically before 25 years of age) but despite this, this condition was not insulin dependent and showed autosomal dominant inheritance [15, 16]. The initial linkage analysis in large families led to discovery of the first MODY gene, encoding glucokinase (GCK) [17, 18]. This was rapidly followed by discovery of genes encoding hepatic nuclear factor 1 alpha (HNF1A) [19], hepatic nuclear factor 4 alpha (HNF4A) [20] and hepatic nuclear factor 1 beta (HNF1B) [21]. Although other genetic causes have subsequently been described, none of these are as common as the four genetic causes initially described [22]. MODY represents between 1.2% and 3.0% of diabetes diagnosed in children, at least in predominately white European populations ([11, 12] and reviewed in [23]).
Discrete clinical features of common MODY subtypes
The discovery of the MODY genes led to the description of discrete clinical courses for different genetic subtypes (Fig. 2). GCK-MODY shows a stable, raised fasting glucose in contrast with the progressive deterioration of glucose over time observed with transcription factor-linked MODY (HNF1A-, HNF4A- and HNF1B-MODY; Fig. 2) [6, 24]. Patients with transcription factor-linked MODY have different associated features based on their underlying aetiology, such as glycosuria in HNF1A-MODY, fetal macrosomia and neonatal hypoglycaemia in HNF4A-MODY, and developmental disorders of the kidney and multiple other organs in HNF1B-MODY (Fig. 2) [6]. The majority of genetic testing requires initial clinical selection followed by molecular genetic testing of either the most likely gene or a panel of all MODY genes [12].
Differential treatment response in MODY subtypes
Probably the most important clinical feature associated with precision diabetes in MODY patients has been the differential treatment response in discrete genetic groups (Fig. 2). GCK-MODY patients do not require treatment [24, 25] and do not respond to either oral agents or low-dose insulin [24, 26]. In contrast, HNF1A- and HNF4A-MODY patients can be treated with low-dose sulfonylureas [6, 27, 28]. Patients who require additional treatment can have dipeptidyl peptidase-4 (DPP-4) inhibitors, glucagon-like peptide-1 (GLP-1) receptor agonist and insulin in addition to sulfonylureas. Patients with HNF1B-MODY require insulin treatment as the response to sulfonylureas and other oral medication is limited [29].
Increasing genetic diagnosis of MODY throughout the world
MODY genetic testing is increasing throughout the world and most developed countries have at least one academic, health service or commercial laboratory providing monogenic diabetes testing. Within the UK, the Exeter laboratory have gone from ∼50 patients being diagnosed with MODY in 1996 to ∼5000 diagnoses in 2016.

Why has precision medicine in monogenic diabetes worked well?

Subgroups that are clearly defined by underlying aetiology

A key feature of monogenic diabetes is that finding a mutation results in diagnosis of a specific subgroup. These specific subcategories have the advantage of being non-overlapping with differential clinical implications (Fig. 3). In addition, for many monogenic diabetes subtypes, the specific mutation will determine the clinical outcome because of genotype–phenotype relationships. For example, in KCNJ11-neonatal diabetes, the mutation severity determines the phenotype [30]. The functional impact of the mutation increases as the phenotype changes from TNDM to isolated PNDM, to PNDM with a neurological phenotype (developmental delay, epilepsy and neonatal diabetes [DEND]) [30, 31]. As another example, HNF4A-MODY patients with the p.R114W mutation (found in ∼15% of HNF4A-MODY patients) have different phenotypes compared with other HNF4A-MODY patients. They showed reduced sensitivity to low-dose sulfonylurea treatment, reduced penetrance and no effect on birthweight [32]. In contrast, GCK-MODY is interesting in that it has a uniform phenotype despite there being considerable functional differences in mutation severity [26]; this is due to compensation by overexpression of the normal allele [33].

Large differences in treatment response in monogenic diabetes

Differences in treatment response can have a large impact in monogenic diabetes. The best example is the enhanced sensitivity to sulfonylureas in HNF1A-MODY, meaning that patients may become severely hypoglycaemic if standard doses are used, and that discontinuing sulfonylureas results in a marked deterioration in blood glucose (a 5% point reduction [31 mmol/mol] in HbA1c) [6, 34]. In a randomised trial sulfonylureas led to a fourfold greater reduction of fasting blood glucose in HNF1A-MODY patients compared with age, BMI and blood glucose level-matched type 2 diabetes patients [27]. This sensitivity to sulfonylureas was initially identified from clinical observation and was not predicted from gene function [34]. In contrast, there is a lack of glycaemic response with oral hypoglycaemic agents or low-dose insulin in patients with GCK-MODY [26]. The lack of efficacy of insulin treatment at a median dose of 0.4 U kg−1 day−1 is also seen in pregnancy as the birthweight of offspring of GCK-MODY patients treated with insulin and without insulin are similar [35]. Insulin is still recommended for individuals with GCK-MODY in some circumstances in pregnancy but even at very high doses, its ability to lower the mother’s blood glucose levels is limited (reviewed in [24]). The lack of response to therapy may be predicted because GCK-MODY patients have a regulated blood glucose that is set at a higher level, as a result of insulin and counter-regulatory hormones being regulated to maintain this elevated glucose level [24, 36].
High-dose sulfonylurea treatment in potassium channel-linked neonatal diabetes (ABCC8- and KCNJ11-neonatal diabetes) had a massive impact on endogenous insulin secretion (measured by C-peptide), which increased from an undetectable level to the level necessary to maintain glucose at near normal values [8]. This resulted in an ∼2 percentage point (22 mmol/mol) improvement in HbA1c in the short term, which persisted for more than 5 years [9, 37]. Importantly, the basis for attempting this therapy arose from the knowledge that the potassium channel is the target of sulfonylureas.

The difficulties of bringing a precision diabetes approach into monogenic diabetes care

Neonatal diabetes: a success story of rapid implementation

The easy clinical recognition of neonatal diabetes combined with a dramatic treatment response following precise genetic diagnosis led to international guidelines being changed within 2 years after gene discovery [38]. The simple clinical guidance was issued that diagnostic genetic testing is required for all patients who developed diabetes before 6 months of age. The simplicity of this guidance greatly helped towards its rapid dissemination worldwide. This was further helped by support from the Wellcome Trust, allowing the Exeter Molecular Genetics Laboratory to offer free rapid comprehensive genetic testing throughout the world for patients with neonatal diabetes until at least 2020. This has resulted in over 1700 patients from 87 countries being tested for neonatal diabetes [7].

MODY: slow uptake into clinical practice

The recognition of MODY has been slow despite MODY being relatively common, the technology for genetic diagnosis being available in most countries and clear treatment recommendations having been defined once a diagnosis is made, both outside and during pregnancy [6, 24]. This is partly because precision diabetes based on genetic testing is a new concept for diabetologists; genetics is not a part of routine clinical training and, traditionally, the speciality of diabetes emphases are based on treatment rather than diagnosis. In addition, a major barrier for the dissemination of precision diabetes in MODY was the lack of single clinical criteria that can accurately identify all MODY patients. MODY cases overlap with type 1 and type 2 diabetes patients with regard to age of onset, BMI, history of parental diabetes, HbA1c levels and treatment (Fig. 4) [39]. The traditional criteria of MODY (diabetes diagnosis <25 years, non-insulin treated and an affected parent) identify only 48% of MODY cases and, hence, are not sufficiently sensitive to be used alone in clinical practice [40]. The net result of these barriers is that the majority of MODY patients are not recognised [40, 41]. In addition, the cost of the genetic test is an important barrier to implementation, although early health economic evidence supports the cost effectiveness of genetic testing for both neonatal diabetes and MODY [42, 43].

A solution to diagnosing MODY when there is no single criterion or threshold

Diagnosing MODY requires a complex multi-dimensional assessment of probability based on more than one clinical criterion. This may be difficult for clinicians but can be easily done by use of a statistical calculator that uses multiple, but readily available, clinical information to assess the probability of a patient having MODY. The ‘MODY Probability Calculator’ was developed by B. Shields and is available without charge at www.​diabetesgenes.​org and on the ‘Diabetes Diagnostic’ app for iOS and Android mobile platforms [39]. In a head-to-head competition, it proved to be as good as clinical experts with more than two decades of experience working with MODY (B. Shields, [University of Exeter Medical School, Exeter, UK] and A. T. Hattersley, personal communication). The probability calculator works best for patients who are not insulin treated. For patients who are insulin treated in whom the diagnosis of MODY is being considered, additional non-genetic tests (islet autoantibody testing and C-peptide analysis) should be considered as ‘rule-out tests’; the presence of islet autoantibodies and/or C-peptide <200 pmol/l effectively rules out MODY [44, 45].
The development of a MODY probability calculator has proved a very promising first step towards precision diabetes. The mobile app is widely used (>6000 downloads to date). This provides a good example of how sophisticated modelling of a complex diagnostic challenge can be simplified into a simple tool that uses readily available clinical information. This approach can greatly help rapid dissemination of precision diabetes.

New technology does not make clinical selection redundant

Next-generation sequencing has transformed our ability to perform genetic testing but it has not removed the need for clinical selection of patients with possible monogenic diabetes for the genetic test. It is now possible to test all genes involved in monogenic diabetes in a single gene panel test, both quickly and efficiently [7, 12]. This gene panel testing approach identifies approximately an additional 25% of monogenic patients with less common causes compared with selected testing of common genetic subtypes [23]. It removes the need to define the likely genetic aetiology/subgroups prior to testing; however, extra care is needed when patients are not selected on phenotype as the prior likelihood of monogenic diabetes is greatly reduced and so false positive findings become more likely [46]. The easy access to sequencing technology has led to laboratories (including commercial laboratories) offering diagnostic testing for monogenic diabetes even when they do not have experience of monogenic diabetes. This has resulted in benign polymorphisms frequently being reported as disease-causing mutations (e.g. 38% of reported cases of HNF1A-MODY in Germany had benign polymorphisms [47]). Clinical selection of patients with potential monogenic diabetes is still required since, even with improved technology, next-generation sequencing cannot find a genetic aetiology in patients who have type 1 or type 2 diabetes.

Precision medicine in type 2 diabetes: comparisons with monogenic diabetes

Difficulty in defining aetiological subgroups in type 2 diabetes

Defining subgroups using molecular genetic testing in type 2 diabetes is very unlikely to result in discrete aetiological subgroups because the genetic predisposition is polygenic rather than monogenic and the clinical phenotype reflects environmental as well as genetic influences (Fig. 3) [48]. We can define aetiological subgroups based on physiological features, such as insulin resistance and beta cell failure. The main problems of using these categories are that these features change over time, there is a lack of agreement on optimum methods of assessment [49] and biochemical assays used in the definitions are not standardised between laboratories [50, 51]. Similar problems are seen in latent autoimmune diabetes in adults (LADA), in which there is a lack of agreement of which islet autoantibodies to study, variation in the assays used to measure a specific antibody and varying thresholds for a positive test [52]. The difficulty in defining subgroups in type 2 diabetes has a major impact on the ability to optimise treatment.

The lack of marked differences in treatment response in type 2 diabetes

In type 2 diabetes, it is unlikely that differences in treatment will be as marked as in monogenic diabetes. On average, most glucose lowering therapies for type 2 diabetes reduce HbA1c by about 1% (11 mmol/mol) [53]. It is known that there is considerable variation in treatment response to glucose lowering therapy in type 2 diabetes but, to date, there has been no description of any subgroups that respond with a dramatic 5 percentage point (31 mmol/mol) change in HbA1c, as observed in individuals with HNF1A-MODY. Pharmacogenetic impacts on treatment responses in type 2 diabetes exist but all have been small to date (<0.5% [5 mmol/mol] HbA1c) [54]. An alternative approach may involve defining type 2 diabetes patients who are unlikely to respond to a specific therapy, with the best example to date being insulin-treated type 2 patients with islet autoantibodies or low C-peptide who do not respond to GLP-1 receptor agonists [55].

An alternative approach for precision medicine in type 2 diabetes

The aim of precision medicine is to find subgroups of patients that have tailored treatment. It does not specify how these subgroups are defined. The successful examples of monogenic diabetes (and also cancer) have been based on molecular genetic analysis but, as outlined above, this is unlikely to be successful in type 2 diabetes and will not be practical to bring into routine clinical practice in primary care. Therefore, rather than defining subgroups based on molecular aetiology, we suggest initial attempts to define subgroups based on differential treatment response to drugs (Fig. 5). The initial analysis for identification of this subgroup should use simple clinical data (e.g. age of diagnosis, sex and BMI) or readily available biomarkers (e.g. eGFR). This approach has the advantage that it can develop relatively quickly because of the use of already available large-scale routine clinical data and subsequently tested using clinical trial data for currently available drugs. The aim will be to create a simple calculator that will use routine clinical information to provide information on the likely HbA1c response and/or risk of adverse effects for available medications. If successful, this approach will be easy to implement in clinical practice and will also be a platform on which future pharmacogenetics or ‘omic’ discoveries can add to [56]. A recent example of the successful implementation of this approach is the use of sex and BMI data for identification of patients with a preferential response to thiazolidinediones (obese female) or sulfonylureas (slim male) [57].

Conclusions

Precision diabetes in monogenic diabetes has been an easy early win but, in contrast, its implementation in type 2 diabetes will be considerably more difficult. Monogenic diabetes has the advantage that there are discrete subgroups that are easily defined by molecular genetics. Frequently, the knowledge of the biology that results from the aetiological gene being identified has helped to define likely treatment response. These early successes have coloured our approach to precision diabetes, favouring genomics-based approaches that search for a single biomarker or a genetic variant with very large effect on treatment response. However, type 2 diabetes is a polygenic condition in which environment, as well as genetic predisposition, play a big role. In this case, an approach concentrating on newer technologies may not be optimum and certainly examining simple clinical criteria like BMI, sex and age should be carried out before rushing to molecular technologies. Finally, one thing that we have learnt from monogenic diabetes, particularly MODY, is that even when there is a clear case, both clinically and economically, for a precision diabetes approach, implementation may be difficult.

Acknowledgements

We would like to thank our many colleagues within the Exeter Monogenic Team, both past and present, and also our numerous collaborators in the UK and internationally. We accept that many of the ideas expressed in this article have come from collective discussion over many years. Some parts of this article were developed from A. T. Hattersley’s EASD/Novo Nordisk Foundation Diabetes Prize for Excellence 2016.

Funding

This work is supported by the MASTERMIND Consortium sponsored by the Medical Research Council (MRC; MR-K005707-1) and by a Wellcome Trust Senior Investigator award given to ATH (and S. Ellard, University of Exeter Medical School, Exeter, UK [WT098395/Z/12/Z]). The work is also supported by the National Institute for Health Research (NIHR) Clinical Research Facility.

Duality of interest

The authors declare that there is no duality of interest associated with this manuscript.

Contribution statement

Both authors were responsible for drafting the article and revising it critically for important intellectual content and approved the version to be published.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

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.

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.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat National Research Council (US) Committee on A Framework for Developing a New Taxonomy of Disease (2011) Toward precision medicine: building a knowledge network for biomedical research and a new taxonomy of disease. National Academies Press, Washington, DC, USA National Research Council (US) Committee on A Framework for Developing a New Taxonomy of Disease (2011) Toward precision medicine: building a knowledge network for biomedical research and a new taxonomy of disease. National Academies Press, Washington, DC, USA
3.
Zurück zum Zitat Iafusco D, Stazi MA, Cotichini R et al (2002) Permanent diabetes mellitus in the first year of life. Diabetologia 45:798–804CrossRefPubMed Iafusco D, Stazi MA, Cotichini R et al (2002) Permanent diabetes mellitus in the first year of life. Diabetologia 45:798–804CrossRefPubMed
4.
Zurück zum Zitat Edghill EL, Dix RJ, Flanagan SE et al (2006) HLA genotyping supports a nonautoimmune etiology in patients diagnosed with diabetes under the age of 6 months. Diabetes 55:1895–1898CrossRefPubMed Edghill EL, Dix RJ, Flanagan SE et al (2006) HLA genotyping supports a nonautoimmune etiology in patients diagnosed with diabetes under the age of 6 months. Diabetes 55:1895–1898CrossRefPubMed
5.
Zurück zum Zitat Rubio-Cabezas O, Flanagan SE, Damhuis A et al (2012) KATP channel mutations in infants with permanent diabetes diagnosed after 6 months of life. Pediatr Diabetes 13:322–325CrossRefPubMed Rubio-Cabezas O, Flanagan SE, Damhuis A et al (2012) KATP channel mutations in infants with permanent diabetes diagnosed after 6 months of life. Pediatr Diabetes 13:322–325CrossRefPubMed
6.
Zurück zum Zitat Murphy R, Ellard S, Hattersley AT (2008) Clinical implications of a molecular genetic classification of monogenic beta-cell diabetes. Nat Clin Pract Endocrinol Metab 4:200–213CrossRefPubMed Murphy R, Ellard S, Hattersley AT (2008) Clinical implications of a molecular genetic classification of monogenic beta-cell diabetes. Nat Clin Pract Endocrinol Metab 4:200–213CrossRefPubMed
7.
Zurück zum Zitat De Franco et al (2015) The effect of early, comprehensive genomic testing on clinical care in neonatal diabetes: an international cohort study. Lancet 386:957–963CrossRefPubMedPubMedCentral De Franco et al (2015) The effect of early, comprehensive genomic testing on clinical care in neonatal diabetes: an international cohort study. Lancet 386:957–963CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Flanagan SE, Haapaniemi E, Russell MA et al (2014) Activating germline mutations in STAT3 cause early-onset multi-organ autoimmune disease. Nat Genet 46:812–814CrossRefPubMedPubMedCentral Flanagan SE, Haapaniemi E, Russell MA et al (2014) Activating germline mutations in STAT3 cause early-onset multi-organ autoimmune disease. Nat Genet 46:812–814CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Pearson ER, Flechtner I, Njølstad PR et al (2006) Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N Engl J Med 355:467–477CrossRefPubMed Pearson ER, Flechtner I, Njølstad PR et al (2006) Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations. N Engl J Med 355:467–477CrossRefPubMed
10.
Zurück zum Zitat Sagen JV, Raeder H, Hathout E et al (2004) Permanent neonatal diabetes due to mutations in KCNJ11 encoding Kir6.2: patient characteristics and initial response to sulfonylurea therapy. Diabetes 53:2713–2718CrossRefPubMed Sagen JV, Raeder H, Hathout E et al (2004) Permanent neonatal diabetes due to mutations in KCNJ11 encoding Kir6.2: patient characteristics and initial response to sulfonylurea therapy. Diabetes 53:2713–2718CrossRefPubMed
11.
Zurück zum Zitat Beltrand J, Elie C, Busiah K et al (2015) Sulfonylurea therapy benefits neurological and psychomotor functions in patients with neonatal diabetes owing to potassium channel mutations. Diabetes Care 38:2033–2041CrossRefPubMed Beltrand J, Elie C, Busiah K et al (2015) Sulfonylurea therapy benefits neurological and psychomotor functions in patients with neonatal diabetes owing to potassium channel mutations. Diabetes Care 38:2033–2041CrossRefPubMed
12.
Zurück zum Zitat Ellard S, Lango Allen H, De Franco E et al (2013) Improved genetic testing for monogenic diabetes using targeted next-generation sequencing. Diabetologia 56:1958–1963CrossRefPubMedPubMedCentral Ellard S, Lango Allen H, De Franco E et al (2013) Improved genetic testing for monogenic diabetes using targeted next-generation sequencing. Diabetologia 56:1958–1963CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Olsen BS, Hahnemann JMD, Schwartz M, Østergaard E (2007) Thiamine-responsive megaloblastic anaemia: a cause of syndromic diabetes in childhood. Pediatr Diabetes 8:239–241CrossRefPubMed Olsen BS, Hahnemann JMD, Schwartz M, Østergaard E (2007) Thiamine-responsive megaloblastic anaemia: a cause of syndromic diabetes in childhood. Pediatr Diabetes 8:239–241CrossRefPubMed
14.
Zurück zum Zitat Nademi Z, Slatter M, Gambineri E et al (2014) Single centre experience of haematopoietic SCT for patients with immunodysregulation, polyendocrinopathy, enteropathy, X-linked syndrome. Bone Marrow Transplant 49:310–312CrossRefPubMed Nademi Z, Slatter M, Gambineri E et al (2014) Single centre experience of haematopoietic SCT for patients with immunodysregulation, polyendocrinopathy, enteropathy, X-linked syndrome. Bone Marrow Transplant 49:310–312CrossRefPubMed
15.
Zurück zum Zitat Tattersall RB (1974) Mild familial diabetes with dominant inheritance. Q J Med 43:339–357PubMed Tattersall RB (1974) Mild familial diabetes with dominant inheritance. Q J Med 43:339–357PubMed
16.
Zurück zum Zitat Tattersall RB, Fajans SS (1975) Prevalence of diabetes and glucose intolerance in 199 offspring of thirty-seven conjugal diabetic parents. Diabetes 24:452–462CrossRef Tattersall RB, Fajans SS (1975) Prevalence of diabetes and glucose intolerance in 199 offspring of thirty-seven conjugal diabetic parents. Diabetes 24:452–462CrossRef
17.
Zurück zum Zitat Froguel P, Vaxillaire M, Sun F et al (1992) Close linkage of glucokinase locus on chromosome 7p to early-onset non-insulin-dependent diabetes mellitus. Nature 356:162–164CrossRefPubMed Froguel P, Vaxillaire M, Sun F et al (1992) Close linkage of glucokinase locus on chromosome 7p to early-onset non-insulin-dependent diabetes mellitus. Nature 356:162–164CrossRefPubMed
18.
Zurück zum Zitat Hattersley AT, Turner RC, Permutt MA et al (1992) Linkage of type 2 diabetes to the glucokinase gene. Lancet 339:1307–1310CrossRefPubMed Hattersley AT, Turner RC, Permutt MA et al (1992) Linkage of type 2 diabetes to the glucokinase gene. Lancet 339:1307–1310CrossRefPubMed
19.
Zurück zum Zitat Yamagata K, Oda N, Kaisaki PJ et al (1996) Mutations in the hepatocyte nuclear factor-1alpha gene in maturity-onset diabetes of the young (MODY3). Nature 384:455–458CrossRefPubMed Yamagata K, Oda N, Kaisaki PJ et al (1996) Mutations in the hepatocyte nuclear factor-1alpha gene in maturity-onset diabetes of the young (MODY3). Nature 384:455–458CrossRefPubMed
20.
Zurück zum Zitat Yamagata K, Furuta H, Oda N et al (1996) Mutations in the hepatocyte nuclear factor-4alpha gene in maturity-onset diabetes of the young (MODY1). Nature 384:458–460CrossRefPubMed Yamagata K, Furuta H, Oda N et al (1996) Mutations in the hepatocyte nuclear factor-4alpha gene in maturity-onset diabetes of the young (MODY1). Nature 384:458–460CrossRefPubMed
21.
Zurück zum Zitat Horikawa Y, Iwasaki N, Hara M et al (1997) Mutation in hepatocyte nuclear factor-1 beta gene (TCF2) associated with MODY. Nat Genet 17:384–385CrossRefPubMed Horikawa Y, Iwasaki N, Hara M et al (1997) Mutation in hepatocyte nuclear factor-1 beta gene (TCF2) associated with MODY. Nat Genet 17:384–385CrossRefPubMed
22.
Zurück zum Zitat McCarthy MI, Hattersley AT (2008) Learning from molecular genetics: novel insights arising from the definition of genes for monogenic and type 2 diabetes. Diabetes 57:2889–2898CrossRefPubMedPubMedCentral McCarthy MI, Hattersley AT (2008) Learning from molecular genetics: novel insights arising from the definition of genes for monogenic and type 2 diabetes. Diabetes 57:2889–2898CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Shepherd M, Shields B, Hammersley S et al (2016) Systematic population screening, using biomarkers and genetic testing, identifies 2.5% of the U.K. pediatric diabetes population with monogenic diabetes. Diabetes Care 39:1879–1888CrossRefPubMedPubMedCentral Shepherd M, Shields B, Hammersley S et al (2016) Systematic population screening, using biomarkers and genetic testing, identifies 2.5% of the U.K. pediatric diabetes population with monogenic diabetes. Diabetes Care 39:1879–1888CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Chakera AJ, Steele AM, Gloyn AL et al (2015) Recognition and management of individuals with hyperglycemia because of a heterozygous glucokinase mutation. Diabetes Care 38:1383–1392CrossRefPubMed Chakera AJ, Steele AM, Gloyn AL et al (2015) Recognition and management of individuals with hyperglycemia because of a heterozygous glucokinase mutation. Diabetes Care 38:1383–1392CrossRefPubMed
25.
Zurück zum Zitat Steele AM, Shields BM, Wensley KJ et al (2014) Prevalence of vascular complications among patients with glucokinase mutations and prolonged, mild hyperglycemia. JAMA 311:279–286CrossRefPubMed Steele AM, Shields BM, Wensley KJ et al (2014) Prevalence of vascular complications among patients with glucokinase mutations and prolonged, mild hyperglycemia. JAMA 311:279–286CrossRefPubMed
26.
Zurück zum Zitat Stride A, Shields B, Gill-Carey O et al (2014) Cross-sectional and longitudinal studies suggest pharmacological treatment used in patients with glucokinase mutations does not alter glycaemia. Diabetologia 57:54–56CrossRefPubMed Stride A, Shields B, Gill-Carey O et al (2014) Cross-sectional and longitudinal studies suggest pharmacological treatment used in patients with glucokinase mutations does not alter glycaemia. Diabetologia 57:54–56CrossRefPubMed
27.
Zurück zum Zitat Pearson ER, Starkey BJ, Powell RJ et al (2003) Genetic cause of hyperglycaemia and response to treatment in diabetes. Lancet 362:1275–1281CrossRefPubMed Pearson ER, Starkey BJ, Powell RJ et al (2003) Genetic cause of hyperglycaemia and response to treatment in diabetes. Lancet 362:1275–1281CrossRefPubMed
28.
Zurück zum Zitat Pearson ER, Pruhova S, Tack CJ et al (2005) Molecular genetics and phenotypic characteristics of MODY caused by hepatocyte nuclear factor 4alpha mutations in a large European collection. Diabetologia 48:878–885CrossRefPubMed Pearson ER, Pruhova S, Tack CJ et al (2005) Molecular genetics and phenotypic characteristics of MODY caused by hepatocyte nuclear factor 4alpha mutations in a large European collection. Diabetologia 48:878–885CrossRefPubMed
29.
Zurück zum Zitat Pearson ER, Badman MK, Lockwood CR et al (2004) Contrasting diabetes phenotypes associated with hepatocyte nuclear factor-1alpha and -1beta mutations. Diabetes Care 27:1102–1107CrossRefPubMed Pearson ER, Badman MK, Lockwood CR et al (2004) Contrasting diabetes phenotypes associated with hepatocyte nuclear factor-1alpha and -1beta mutations. Diabetes Care 27:1102–1107CrossRefPubMed
30.
Zurück zum Zitat Hattersley AT, Ashcroft FM (2005) Activating mutations in Kir6.2 and neonatal diabetes: new clinical syndromes, new scientific insights, and new therapy. Diabetes 54:2503–2513CrossRefPubMed Hattersley AT, Ashcroft FM (2005) Activating mutations in Kir6.2 and neonatal diabetes: new clinical syndromes, new scientific insights, and new therapy. Diabetes 54:2503–2513CrossRefPubMed
31.
Zurück zum Zitat Gloyn AL, Diatloff-Zito C, Edghill EL et al (2006) KCNJ11 activating mutations are associated with developmental delay, epilepsy and neonatal diabetes syndrome and other neurological features. Eur J Hum Genet 14:824–830CrossRefPubMed Gloyn AL, Diatloff-Zito C, Edghill EL et al (2006) KCNJ11 activating mutations are associated with developmental delay, epilepsy and neonatal diabetes syndrome and other neurological features. Eur J Hum Genet 14:824–830CrossRefPubMed
32.
Zurück zum Zitat Laver TW, Colclough K, Shepherd M et al (2016) The common p.R114W HNF4A mutation causes a distinct clinical subtype of monogenic diabetes. Diabetes 65:3212–3217CrossRefPubMedPubMedCentral Laver TW, Colclough K, Shepherd M et al (2016) The common p.R114W HNF4A mutation causes a distinct clinical subtype of monogenic diabetes. Diabetes 65:3212–3217CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Sturis J, Kurland IJ, Byrne MM et al (1994) Compensation in pancreatic beta-cell function in subjects with glucokinase mutations. Diabetes 43:718–723CrossRefPubMed Sturis J, Kurland IJ, Byrne MM et al (1994) Compensation in pancreatic beta-cell function in subjects with glucokinase mutations. Diabetes 43:718–723CrossRefPubMed
34.
Zurück zum Zitat Pearson ER, Liddell WG, Shepherd M et al (2000) Sensitivity to sulphonylureas in patients with hepatocyte nuclear factor-1alpha gene mutations: evidence for pharmacogenetics in diabetes. Diabet Med 17:543–545CrossRefPubMed Pearson ER, Liddell WG, Shepherd M et al (2000) Sensitivity to sulphonylureas in patients with hepatocyte nuclear factor-1alpha gene mutations: evidence for pharmacogenetics in diabetes. Diabet Med 17:543–545CrossRefPubMed
35.
Zurück zum Zitat Spyer G, Macleod KM, Shepherd M et al (2009) Pregnancy outcome in patients with raised blood glucose due to a heterozygous glucokinase gene mutation. Diabet Med 26:14–18CrossRefPubMed Spyer G, Macleod KM, Shepherd M et al (2009) Pregnancy outcome in patients with raised blood glucose due to a heterozygous glucokinase gene mutation. Diabet Med 26:14–18CrossRefPubMed
36.
Zurück zum Zitat Guenat E, Seematter G, Philippe J et al (2000) Counterregulatory responses to hypoglycemia in patients with glucokinase gene mutations. Diabetes Metab 26:377–384PubMed Guenat E, Seematter G, Philippe J et al (2000) Counterregulatory responses to hypoglycemia in patients with glucokinase gene mutations. Diabetes Metab 26:377–384PubMed
37.
Zurück zum Zitat Iafusco D, Bizzarri C, Cadario F et al (2011) No beta cell desensitisation after a median of 68 months on glibenclamide therapy in patients with KCNJ11-associated permanent neonatal diabetes. Diabetologia 54:2736–2738CrossRefPubMed Iafusco D, Bizzarri C, Cadario F et al (2011) No beta cell desensitisation after a median of 68 months on glibenclamide therapy in patients with KCNJ11-associated permanent neonatal diabetes. Diabetologia 54:2736–2738CrossRefPubMed
38.
Zurück zum Zitat Hattersley A, Bruining J, Shield J et al (2006) ISPAD clinical practice consensus guidelines 2006-2007. The diagnosis and management of monogenic diabetes in children. Pediatr Diabetes 7:352–360CrossRefPubMed Hattersley A, Bruining J, Shield J et al (2006) ISPAD clinical practice consensus guidelines 2006-2007. The diagnosis and management of monogenic diabetes in children. Pediatr Diabetes 7:352–360CrossRefPubMed
39.
Zurück zum Zitat Shields BM, McDonald TJ, Ellard S et al (2012) The development and validation of a clinical prediction model to determine the probability of MODY in patients with young-onset diabetes. Diabetologia 55:1265–1272CrossRefPubMedPubMedCentral Shields BM, McDonald TJ, Ellard S et al (2012) The development and validation of a clinical prediction model to determine the probability of MODY in patients with young-onset diabetes. Diabetologia 55:1265–1272CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Shields BM, Hicks S, Shepherd MH et al (2010) Maturity-onset diabetes of the young (MODY): how many cases are we missing? Diabetologia 53:2504–2508CrossRefPubMed Shields BM, Hicks S, Shepherd MH et al (2010) Maturity-onset diabetes of the young (MODY): how many cases are we missing? Diabetologia 53:2504–2508CrossRefPubMed
41.
Zurück zum Zitat Pihoker C, Gilliam LK, Ellard S et al (2013) Prevalence, characteristics and clinical diagnosis of maturity onset diabetes of the young due to mutations in HNF1A, HNF4A, and glucokinase: results from the SEARCH for Diabetes in Youth. J Clin Endocrinol Metab 98:4055–4062CrossRefPubMedPubMedCentral Pihoker C, Gilliam LK, Ellard S et al (2013) Prevalence, characteristics and clinical diagnosis of maturity onset diabetes of the young due to mutations in HNF1A, HNF4A, and glucokinase: results from the SEARCH for Diabetes in Youth. J Clin Endocrinol Metab 98:4055–4062CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Naylor RN, John PM, Winn AN et al (2014) Cost-effectiveness of MODY genetic testing: translating genomic advances into practical health applications. Diabetes Care 37:202–209CrossRefPubMed Naylor RN, John PM, Winn AN et al (2014) Cost-effectiveness of MODY genetic testing: translating genomic advances into practical health applications. Diabetes Care 37:202–209CrossRefPubMed
43.
Zurück zum Zitat Greeley SAW, John PM, Winn AN et al (2011) The cost-effectiveness of personalized genetic medicine: the case of genetic testing in neonatal diabetes. Diabetes Care 34:622–627CrossRefPubMedPubMedCentral Greeley SAW, John PM, Winn AN et al (2011) The cost-effectiveness of personalized genetic medicine: the case of genetic testing in neonatal diabetes. Diabetes Care 34:622–627CrossRefPubMedPubMedCentral
44.
Zurück zum Zitat McDonald TJ, Colclough K, Brown R et al (2011) Islet autoantibodies can discriminate maturity-onset diabetes of the young (MODY) from Type 1 diabetes. Diabet Med 28:1028–1033CrossRefPubMed McDonald TJ, Colclough K, Brown R et al (2011) Islet autoantibodies can discriminate maturity-onset diabetes of the young (MODY) from Type 1 diabetes. Diabet Med 28:1028–1033CrossRefPubMed
45.
Zurück zum Zitat Besser REJ, Shepherd MH, McDonald TJ et al (2011) Urinary C-peptide creatinine ratio is a practical outpatient tool for identifying hepatocyte nuclear factor 1-/hepatocyte nuclear factor 4- maturity-onset diabetes of the young from long-duration type 1 diabetes. Diabetes Care 34:286–291CrossRefPubMedPubMedCentral Besser REJ, Shepherd MH, McDonald TJ et al (2011) Urinary C-peptide creatinine ratio is a practical outpatient tool for identifying hepatocyte nuclear factor 1-/hepatocyte nuclear factor 4- maturity-onset diabetes of the young from long-duration type 1 diabetes. Diabetes Care 34:286–291CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Flannick J, Beer NL, Bick AG et al (2013) Assessing the phenotypic effects in the general population of rare variants in genes for a dominant Mendelian form of diabetes. Nat Publ Group 45:1380–1385 Flannick J, Beer NL, Bick AG et al (2013) Assessing the phenotypic effects in the general population of rare variants in genes for a dominant Mendelian form of diabetes. Nat Publ Group 45:1380–1385
47.
Zurück zum Zitat Awa WL, Thon A, Raile K et al (2011) Genetic and clinical characteristics of patients with HNF1A gene variations from the German-Austrian DPV database. Eur J Endocrinol 164:513–520CrossRefPubMed Awa WL, Thon A, Raile K et al (2011) Genetic and clinical characteristics of patients with HNF1A gene variations from the German-Austrian DPV database. Eur J Endocrinol 164:513–520CrossRefPubMed
49.
Zurück zum Zitat Borai A, Livingstone C, Ferns GAA (2007) The biochemical assessment of insulin resistance. Ann Clin Biochem 44:324–342CrossRefPubMed Borai A, Livingstone C, Ferns GAA (2007) The biochemical assessment of insulin resistance. Ann Clin Biochem 44:324–342CrossRefPubMed
50.
Zurück zum Zitat Parfitt C, Church D, Armston A et al (2015) Commercial insulin immunoassays fail to detect commonly prescribed insulin analogues. Clin Biochem 48:1354–1357CrossRefPubMed Parfitt C, Church D, Armston A et al (2015) Commercial insulin immunoassays fail to detect commonly prescribed insulin analogues. Clin Biochem 48:1354–1357CrossRefPubMed
51.
Zurück zum Zitat Little RR, Rohlfing CL, Tennill AL et al (2008) Standardization of C-peptide measurements. Clin Chem 54:1023–1026CrossRefPubMed Little RR, Rohlfing CL, Tennill AL et al (2008) Standardization of C-peptide measurements. Clin Chem 54:1023–1026CrossRefPubMed
52.
Zurück zum Zitat Tuomi T, Santoro N, Caprio S et al (2014) The many faces of diabetes: a disease with increasing heterogeneity. Lancet 383:1084–1094CrossRefPubMed Tuomi T, Santoro N, Caprio S et al (2014) The many faces of diabetes: a disease with increasing heterogeneity. Lancet 383:1084–1094CrossRefPubMed
53.
Zurück zum Zitat Bennett WL, Maruthur NM, Singh S et al (2011) Comparative effectiveness and safety of medications for type 2 diabetes: an update including new drugs and 2-drug combinations. Ann Intern Med 154:602–613CrossRefPubMedPubMedCentral Bennett WL, Maruthur NM, Singh S et al (2011) Comparative effectiveness and safety of medications for type 2 diabetes: an update including new drugs and 2-drug combinations. Ann Intern Med 154:602–613CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Dawed AY, Zhou K, Pearson ER (2016) Pharmacogenetics in type 2 diabetes: influence on response to oral hypoglycemic agents. Pharm Pers Med 9:17–29 Dawed AY, Zhou K, Pearson ER (2016) Pharmacogenetics in type 2 diabetes: influence on response to oral hypoglycemic agents. Pharm Pers Med 9:17–29
55.
Zurück zum Zitat Jones AG, McDonald TJ, Shields BM et al (2016) Markers of β-cell failure predict poor glycemic response to GLP-1 receptor agonist therapy in type 2 diabetes. Diabetes Care 39:250–257PubMed Jones AG, McDonald TJ, Shields BM et al (2016) Markers of β-cell failure predict poor glycemic response to GLP-1 receptor agonist therapy in type 2 diabetes. Diabetes Care 39:250–257PubMed
56.
Zurück zum Zitat Zeevi D, Korem T, Zmora N et al (2015) Personalized nutrition by prediction of glycemic responses. Cell 163:1079–1094CrossRefPubMed Zeevi D, Korem T, Zmora N et al (2015) Personalized nutrition by prediction of glycemic responses. Cell 163:1079–1094CrossRefPubMed
57.
Zurück zum Zitat Shields BM, Longergan M, Dennis J et al (2015) Patient characteristics are associated with treatment response to second line glucose lowering therapy: a MASTERMIND study abstracts of 51st EASD annual meeting. Diabetologia 58(Suppl 1):S405 Shields BM, Longergan M, Dennis J et al (2015) Patient characteristics are associated with treatment response to second line glucose lowering therapy: a MASTERMIND study abstracts of 51st EASD annual meeting. Diabetologia 58(Suppl 1):S405
Metadaten
Titel
Precision diabetes: learning from monogenic diabetes
verfasst von
Andrew T. Hattersley
Kashyap A. Patel
Publikationsdatum
17.03.2017
Verlag
Springer Berlin Heidelberg
Erschienen in
Diabetologia / Ausgabe 5/2017
Print ISSN: 0012-186X
Elektronische ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-017-4226-2

Weitere Artikel der Ausgabe 5/2017

Diabetologia 5/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

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