The concept of precision medicine is becoming increasingly popular. The use of big data, genomics and other “omics” like metabolomics, proteomics and transcriptomics could make the dream of personalised medicine become a reality in the near future. As far as polygenic forms of diabetes like type 2 and type 1 diabetes are concerned, interesting leads are emerging, but precision diabetes is still in its infancy. However, with regard to monogenic forms of diabetes like maturity-onset diabetes of the young and neonatal diabetes mellitus, rapid strides have been made and precision diabetes has already become part of the clinical tools used at advanced diabetes centres. In patients with some monogenic form of diabetes, if the appropriate gene defects are identified, insulin injections can be stopped and be replaced by oral sulphonylurea drugs. In the coming years, rapid advances can be expected in the field of precision diabetes, thereby making the control of diabetes more effective and hopefully leading to prevention of its complications and improvement of the quality of life of people afflicted with diabetes.

  • Till recently, a “one-size-fits-all” approach was used to treat most forms of diabetes. With the availability of big data and analytic tools like genomics, epigenetics, metabolomics and proteomics, precision diabetes is now slowly becoming a reality. Precision diabetes is already being applied to monogenic forms of diabetes like maturity-onset diabetes of the young, neonatal diabetes mellitus and congenital hyperinsulinaemic hypoglycaemia. In many such cases, this can be a life changer, as insulin can be stopped completely and be replaced with oral glucose-lowering agents. In other forms of diabetes like type 2 diabetes, precision diabetes promises to be of use in personalised therapy for people affected with the disorder.

The National Institutes of Health (NIH), USA, define precision medicine as an emerging approach to disease treatment and prevention that takes into account individual variability in genes, environment and lifestyle [1]. This concept is not new, and in the past, patients were prescribed optimal treatment by clinicians based on the available data and resources. In its crudest sense, conducting blood transfusion based on the specific blood group of a recipient is an example of precision medicine [2]. The crux of precision medicine is custom delivery of healthcare, with tailored treatment at the level of the individual patient. However, in spite of extensive epidemiological and physiological characterisation and elucidating pathophysiological pathways, we are still far away from predicting prognoses, or even selecting the most effective medicines for a given patient. Most therapeutic approaches are based on population averages, which unfortunately do not fit -everyone [3].

Since these current methods of disease categorisation are outmoded and have not achieved their objectives, a new concept has arisen in the recent years. It is recognised that individuals who share the same diagnostic labels may have very heterogeneous presentations and clinical courses and may respond differently to therapeutic interventions. This has led to the concept of personalised medicine, which, when further refined by scientific inputs, -becomes precision medicine [4].

Precision medicine refers to collecting specific information about a patient and prescribing a specific treatment. This involves defining disease subtypes and biomarkers that can identify patients who are likely to benefit from a specific treatment or those who are likely to experience a particular side effect. Application of genotypic and phenotypic information forms the basis of precision medicine [5]. Personalised medicine not only incorporates the principles of precision medicine but also takes into account patients’ preferences and social context, as well as other factors such as affordability of ther-apies.

One of the important reasons for the rapid development of precision medicine in recent years is the availability of big data and the advancement of technologies to mine these data [6, 7]. It is believed that traditional sources of medical information, such as patient history, physical examination and laboratory workup, when augmented by data mining of deep phenotypic data and genomic analyses coupled with epigenetic, metabolomic, proteomic and transcriptomic data, can help deliver precision medicine to individual patients [4, 8].

A plethora of transformative and disruptive technologies such as DNA sequencing, functional investigation, the emerging field of epigenetics, genome-wide association studies and miRNA scans, as well as a range of “omic” measurements, can help in the detailed in vivo dissection of physiology and pathology, and this improves diagnosis and treatment. They can also provide the basis for refinement of disease subtypes and support the effective optimisation of disease management to match an individual pathology or genetic make-up [9]. The field of oncology has been one of the first to adopt precision medicine, but other fields, including endocrinology, are catching up fast.

Diabetes mellitus broadly encompasses four categories, namely, type 1 diabetes mellitus (T1DM), type 2 diabetes mellitus (T2DM), monogenic diabetes and gestational diabetes.

T1DM is an autoimmune disease with progressive destruction of β-cells, leading to polyuria, polydipsia, weight loss and hyperglycaemia. T1DM requires lifelong administration of insulin injections several times a day as endogenous insulin secretion becomes near zero. Making a proper diagnosis of T1DM with clear evidence of β-cell destruction through the presence of islet cell autoantibodies and demonstration of absent β-cell function as measured by levels of C peptide and prescribing the appropriate doses of insulin are the basis for precision medicine in the case of T1DM, although some heterogeneity exists even here.

T2DM is a heterogeneous group of disorders predominantly due to insulin resistance and some degree of an insulin secretion defect. T2DM has a range of risk factors, aetiologies and clinical presentations.

There are also several forms of monogenic diabetes, which are caused by highly penetrant single gene defects; the three major types maturity-onset diabetes of the young (MODY), neonatal diabetes mellitus (NDM) and congenital hyperinsulinaemic hypoglycaemia (CHH) are the most common. It is with these forms of diabetes that precision diabetes has already come to be used in the clinical setting [10].

This review will first describe the role of precision medicine in T2DM and then deal with the monogenic forms of diabetes.

Defining subgroups using molecular testing is much more difficult in T2DM, as it is a polygenic disorder and the clinical phenotype reflects environmental as well as genetic influences [11]. One approach for precision medicine in T2DM is to categorise patients based on differential responses to drugs, and to then investigate the molecular underpinnings of each subgroup using next-generation sequencing platforms and gene arrays. In clinical practice this approach is not easy to implement, although pharmacogenomic developments continue to be made [12].

In contrast to monogenic diabetes, where the gene mutation is causal and has a high phenotypic correlation, T2DM involves multiple, common, low-impact risk variants. The emerging availability of genomic and electronic health data from large populations, the so-called big data, is a powerful tool for bringing precision medicine to T2DM [13, 14]. For example, recently, at least five clusters of T2DM with differing clinical features have been identified [15].

A common nonsense mutation in the TBC1D4 gene that substantially increases the risk of T2DM was recently identified in a Greenlandic Inuit population [16]. This variant is shown to exclusively increase postprandial glucose levels. The frequency and effect of the TBC1D4 mutation on glucose metabolism and T2DM were determined in two related populations: Canadian Inuit and Alaskan Inuit [17]. The mutation in the TBC1D4 gene is common among North American Inuit, and results in elevated postprandial glucose levels and an underdiagnosis of T2DM unless an oral glucose tolerance test is performed. This is one example of how precision medicine may be applied at the population level.

Some pharmacogenomic advances have been made in relation to antidiabetic drugs. Variations in the cytochrome P450 (CYP) enzymes contribute to oral antidiabetic drug metabolism in the liver and affect drug disposition and efficacy. Genetic variants in CYP2C9*2 (I359L) and CYP2C9*3 (R114C) have been shown to be associated with reduced blood sulphonylurea (SU) clearance [18, 19]. Furthermore, variants in CYP2C8 were found to influence the efficacy of repaglinide and rosiglitazone [20].

Various studies have shown that rs12208357, rs34130495, rs34059508 and rs72552763 are related to the SLC22A1 gene, which encodes organic cation transporter 1, and are genetic markers for the efficacy and excretion of metformin [21-24].

The SNP rs11212617, which is located near the ATM locus, was found to be associated with HbA1c levels in response to metformin in a large-scale genome-wide association study conducted in European T2DM populations [25]. One study has demonstrated that the PAX4 variants rs6367136 and rs10229583 and the PSMD6 variant rs831571 were correlated with the therapeutic efficacy of repaglinide and rosiglitazone in Chinese T2DM patients [26, 27].

Dujic et al. [28] demonstrated the impact of genetic factors on gastrointestinal tolerance to metformin. The Genetics of Diabetes Audit and Research in Tayside Scotland (GoDARTS) study [29] had also shown that reduced-function alleles of the OCT1 gene are associated with increased intolerance to metformin. Recently, it has been suggested that serotonin reuptake transporter might also be involved in intestinal metformin absorption. The number of low-expressing serotonin reuptake transporter alleles significantly increased the odds of metformin tolerance. These results suggest that gastrointestinal side effects of metformin could be related to the reduced uptake of intestinal serotonin [23].

Another pharmacogenetic approach to treatment response in T2DM was demonstrated with the use of thiazolidinediones (TZDs), compounds that are transported into the liver by OATP1B1 (encoded by the SLCO1B1 gene) and metabolised by the CYP450 2C8 enzyme (encoded by the CYP2C8 gene). Although variants in the -CYP2C8 gene (the CYP2C8*3 allele) have been shown to alter TZD pharmacokinetics, the CYP2C8*3 allele has not been shown to alter its efficacy. In an elegant study [30], 833 patients with T2DM treated with pioglitazone/rosiglitazone were genotyped for CYP2C8 and SLCO1B1 functional variants. The CYP2C8*3 variant was significantly associated with reduced glycaemic response to rosiglitazone and less weight gain, whereas the SLCO1B1 521T>C variant was associated with enhanced glycaemic response to rosiglitazone. Patients with both genotypes (super-responders) had a significantly greater HbA1c reduction. Interestingly, neither of the variants had a significant impact on pioglitazone response. This highlights the importance of studying transporter and metabolising genes as a predictor of treatment response by identifying those individuals who can benefit from the therapeutic advantages of TZDs.

There are also reports showing that genetic polymorphisms, such as TCF7L2 polymorphisms, may influence the response to dipeptidyl peptidase-4 (DPP-4) inhibitors [31-33]. There are also some studies suggesting that gene polymorphisms may mediate the response to GLP-1 (glucagon-like peptide-1) receptor agonists [34] and DPP-4 inhibitors [35].

A multitude of newer antidiabetic agents are now available. These include DPP-4 inhibitors, sodium/glucose cotransporter 2 (SGLT2) inhibitors and GLP-1 receptor agonists. It is now known that there are at least 8 pathophysiological defects in T2DM. These drugs help address several of the defects. It is also known that there could be ethnic differences in the response to these drugs; for example, the DPP-4 drugs seem to work better in Asians than in Europeans [36-40]. This could be because β-cell dysfunction is more severe in Asians, particularly in Indians [41]. Indeed, it has been shown that Asian Indians and Pima Indians represent two extreme phenotypes of T2DM, one leaner with more insulin deficiency and the other obese with severe insulin resistance [42]. This suggests that an insulin secretagogue would be preferable for the former and insulin sensitisers for the latter. This is one sample application of personalised or precision medicine at the population level.

In a brilliant review, Schwartz et al. [43] make out a case for a new classification system of diabetes and discuss the rationale for and implications of the β-cell-centred classification schema for T2DM. They also emphasise the concept of epigenetics and inflammation, as well as the role of inflammation and immune dysregulation, in the pathophysiology of T2DM [44]. They suggest how data from “multiomics” could be integrated with electronic medical records to provide “dynamic decision support” for recommendations regarding the treatment of T2DM. They envisage that in the not-too-distant future, data on genomic, epigenomic, proteomic and metabolomic markers could be available on a chip. This will help to classify individuals with diabetes more accurately, and thus make diabetes treatment more patient centred and precision medicine in diabetes a reality.

Figure 1 summarises the concept of a futuristic model of precision diabetes which could be applied at a diabetes clinic to offer personalised diabetes care [44].

Fig. 1.

Precision diabetes: a futuristic model. Adapted from Schwartz et al. [44].

Fig. 1.

Precision diabetes: a futuristic model. Adapted from Schwartz et al. [44].

Close modal

In contrast to the polygenic forms of diabetes like T2DM and T1DM, where precision diabetes is still an evolving field, there are many forms of monogenic diabetes where precision diabetes has already found a place in clinical practice. We will deal with three of these conditions: (1) NDM, (2) CHH, and (3) MODY.

Neonatal Diabetes Mellitus

NDM is defined as diabetes which develops in neonates under 6 months of age. About 50% of the patients with NDM have mutations in the KATP channel genes, namely, KCNJ11 and ABCC8 [45]. Patients with mutations in these two genes respond to high-dose SUs and show excellent glucose control without any increase in hypoglycaemia and glucose variability [46]. Improvements in neurological function are also seen in these patients after treatment with SU [47, 48]. In such patients, treatment with insulin reduces the glucose level but does not elicit an optimal response – and, more specifically, the neurological functions do not show any improvement [49]. SUs close the SU channel in the cells so that exocytosis of insulin takes place, thereby correcting the defect. Since KATP channels are present in β-cells, the brain, the liver, etc., all cells are corrected simultaneously, thus improving their functions as well. This is an excellent illustration of precision diabetes and one of the most exciting clinical situations in diabetology. Switching a child aged just a few weeks or months to oral tablets, from what would otherwise have meant lifelong insulin injections, is nothing short of a miracle to the child and the family, and we have been able to help many such children [50].

Not all mutations in the KCNJ11 and ABCC8 genes, however, lead to NDM. Only some mutations are clinically actionable. This needs to be investigated by functional methods and through segregation analysis. The knowledge of causal mutations has helped in the successful transfer of children from insulin to SUs, as shown in studies by Hattersley and Ashcroft [51] and recently by our group [52]. Patients with transient neonatal diabetes due to 6q24 methylation abnormalities can be treated with low-dose SU when they relapse, while patients with other subtypes of neonatal diabetes require insulin treatment [53]. The ability to explain additional clinical abnormalities that are associated with the underlying genetic cause is one of the major benefits of precision medicine. One such example is cardiac defects in patients with GATA6 mutations and remission of transient diabetes in patients with 6q24 methylation abnormalities [54].

Patients with transient neonatal diabetes due to 6q24methylation abnormalities can be treated with low-dose SUs [55] when they relapse, whereas patients with NDM due to KATP channel mutations require high-dose SU and those with non-KATP channel mutations require insulin treatment [56].

Congenital Hyperinsulinaemic Hypoglycaemia

Another important area where precision diabetes is playing a role is CHH. Here, hypoglycaemia (low blood glucose) manifests just after birth or in the first few months of life. This hypoglycaemia can be severe, debilitating and persistent. In many of these neonates with hypoglycaemia, definite mutations can be identified that are responsible for the condition. We reported on a series of CHH cases from our centre [57]. Some of these neonates respond to diazoxide, while others require subtotal pancreatectomy. Before such a drastic procedure, it is useful to confirm the diagnosis by genetic testing, and this is where precision diabetes plays a role [57].

In the case of conditions such as thiamine responsive megaloblastic anaemia syndrome, an early genetic diagnosis and treatment with thiamine help in the control of diabetes [58]. For IPEX and some severe monogenic autoimmune syndromes, a genetic diagnosis helps in early detection and allows consideration of early curative bone marrow transplantation before patients become severely affected [59].

Maturity-Onset Diabetes of the Young

By its classic definition, MODY presents in non-obese individuals before the age of 25 years, is inherited in an autosomal dominant manner, is primarily due to β-cell dysfunction and can be treated without insulin for at least 5 years [60]. Since the presentation of MODY has characteristics of both type 1 (early onset, lean) and type 2 (family history of diabetes, with β-cell function), it is often misdiagnosed. After the discovery of the MODY genes, a genetic workup helps in classifying MODY accurately. This is very important, since some of the MODY subtypes entail specific therapeutic recommendations based on the genetic aetiology. The transcription factor genes HNF1A (MODY 3) and HNF4A (MODY 1) promote transcription of multiple genes related to glucose metabolism, -insulin secretion and insulin production. Diagnosis of MODY 1 and 3 is important for clinical therapy, as patients with those types are very sensitive to SUs [61-67]. This is due to decreased expression of the target genes HNF1A and 4A in the liver, leading to decreased uptake of SUs, in turn resulting in increased circulating levels of SUs [68]. Therefore, HNF1A and 4A MODY patients need lower doses of SUs, which are the first-line treatment for MODY 1 and 3 [69]. It has been demonstrated that switching of treatment from insulin to SU improved HbA1c levels in MODY 3 and MODY 1 patients. In a randomised trial, Pearson et al. [70] showed that SUs lead to a 4-fold greater reduction of fasting plasma glucose levels in HNF1A MODY patients compared to T2DM patients matched for age, BMI and blood glucose levels.

GCK MODY (MODY 2) patients do not require insulin or oral drugs, as these patients have a regulated blood glucose set at a higher threshold level [71, 72]. They respond to diet and exercise and rarely develop complications from diabetes. Only during pregnancy do GCK MODY women need insulin [73]. However, its validation in non-European populations has not been tested.

In Asian Indians, T2DM occurs earlier and often overlaps with MODY, and hence understanding and recognising MODY subtypes in this population gains more importance. Our work has revealed important insights into the molecular genetics of MODY by showing the presence of novel mutations in the HNF1A (MODY 3) [74] and HNF4A (MODY 1) [61] genes. We showed that about 9% of clinically diagnosed MODY subjects had MODY 3 [75], while 3% had MODY 1. We also identified MODY 5 in some of our patients [76]. We have recently reported on a comprehensive analysis of all 14 MODY genes and have also reported on what is likely to be a new MODY gene, with a mutation in the NKX6-1 gene, located on chromosome 7 [77].

Table 1 summarises the usefulness of precision diabetes in monogenic forms of diabetes. Advancements in genetic testing due to the development of next-generation sequencing have made comprehensive analyses of all known genetic bases for monogenic diabetes possible. This technology has also helped in reducing the turnaround time for genetic testing. A rapid and precise genetic diagnosis is now possible even before the development of all clinical features [49, 51]. Figure 2 summarises the advantages of precision diabetes.

Table 1.

Usefulness of precision diabetes in monogenic diabetes

Usefulness of precision diabetes in monogenic diabetes
Usefulness of precision diabetes in monogenic diabetes
Fig. 2.

Advantages of precision diabetes -applications.

Fig. 2.

Advantages of precision diabetes -applications.

Close modal

Rapid strides are being made in the field of precision diabetes. While it is still too early to say that we have reached a stage where every patient with diabetes can be given a personalised form of treatment, we are beginning to understand that one size does not fit all. In the case of T2DM, clustering of the disorder has now been identified as the first step in precision diabetes. In the case of monogenic forms of diabetes, particularly in MODY, NDM and CHH, genetic testing has come to stay and can be life-changing for those individuals with certain genetic variants. In the years to come, precision diabetes can be expected to play a big role in improving the quality of life of people with all forms of diabetes and help them achieve better control of their diabetes and thereby prevent complications arising from the disorder.

Our work on Precision Diabetes is supported by the INSPIRED project, a grant from NIHR, UK. Financial support for the Monogenic Diabetes study by ICMR, India, is gratefully acknowledged. We also thank ICMR for support from the ICMR Center for Advanced Research (CAR) on Diabetes.

1.
NIH definition.
https://ghr.nlm.nih.gov/primer/precisionmedicine/definition. Accessed on December 22, 2018
2.
Bayne-Jones
S
.
Dr. Karl Landstteiner Nobel Prize Laureate in Medicine, 1930
.
Science
.
1931
Jun
;
73
(
1901
):
599
604
.
[PubMed]
0036-8075
3.
Florez
JC
.
Precision Medicine in Diabetes: is it time?
Diabetes Care
.
2016
Jul
;
39
(
7
):
1085
8
.
[PubMed]
0149-5992
4.
Mirnezami
R
,
Nicholson
J
,
Darzi
A
.
Preparing for precision medicine
.
N Engl J Med
.
2012
Feb
;
366
(
6
):
489
91
.
[PubMed]
0028-4793
5.
Klonoff
DC
.
Precision medicine for managing diabetes
.
J Diabetes Sci Technol
.
2015
Jan
;
9
(
1
):
3
7
.
[PubMed]
1932-2968
6.
Khan
N
,
Yaqoob
I
,
Hashem
IA
,
Inayat
Z
,
Ali
WK
,
Alam
M
, et al.
Big data: survey, technologies, opportunities, and challenges
.
ScientificWorldJournal
.
2014
;
2014
:
712826
.
[PubMed]
2356-6140
7.
Klonoff
DC
.
Twelve modern digital technologies that are transforming decision making for diabetes and all areas of health care
.
J Diabetes Sci Technol
.
2013
Mar
;
7
(
2
):
291
5
.
[PubMed]
1932-2968
8.
Ashley
EA
.
Towards precision medicine
.
Nat Rev Genet
.
2016
Aug
;
17
(
9
):
507
22
.
[PubMed]
1471-0056
9.
McCarthy
MI
.
Painting a new picture of personalised medicine for diabetes
.
Diabetologia
.
2017
May
;
60
(
5
):
793
9
.
[PubMed]
0012-186X
10.
Shepherd
M
,
Shields
B
,
Ellard
S
,
Rubio-Cabezas
O
,
Hattersley
AT
.
A genetic diagnosis of HNF1A diabetes alters treatment and improves glycaemic control in the majority of insulin-treated patients
.
Diabet Med
.
2009
Apr
;
26
(
4
):
437
41
.
[PubMed]
0742-3071
11.
Florez
JC
.
Pharmacogenetics in type 2 diabetes: precision medicine or discovery tool?
Diabetologia
.
2017
May
;
60
(
5
):
800
7
.
[PubMed]
0012-186X
12.
Huang
C
,
Florez
JC
.
Pharmacogenetics in type 2 diabetes: potential implications for clinical practice
.
Genome Med
.
2011
Nov
;
3
(
11
):
76
.
[PubMed]
1756-994X
13.
Klonoff
DC
.
Personalized medicine for diabetes
.
J Diabetes Sci Technol
.
2008
May
;
2
(
3
):
335
41
.
[PubMed]
1932-2968
14.
Maruthur
NM
,
Gribble
MO
,
Bennett
WL
,
Bolen
S
,
Wilson
LM
,
Balakrishnan
P
, et al.
The pharmacogenetics of type 2 diabetes: a systematic review
.
Diabetes Care
.
2014
;
37
(
3
):
876
86
.
[PubMed]
0149-5992
15.
Ahlqvist
E
,
Storm
P
,
Käräjämäki
A
,
Martinell
M
,
Dorkhan
M
,
Carlsson
A
, et al.
Novel subgroups of adult-onset diabetes and their association with outcomes: a data-driven cluster analysis of six variables
.
Lancet Diabetes Endocrinol
.
2018
May
;
6
(
5
):
361
9
.
[PubMed]
2213-8587
16.
Dash
S
,
Sano
H
,
Rochford
JJ
,
Semple
RK
,
Yeo
G
,
Hyden
CS
, et al.
A truncation mutation in TBC1D4 in a family with acanthosis nigricans and postprandial hyperinsulinemia
.
Proc Natl Acad Sci USA
.
2009
Jun
;
106
(
23
):
9350
5
.
[PubMed]
0027-8424
17.
Manousaki
D
,
Kent
JW
 Jr
,
Haack
K
,
Zhou
S
,
Xie
P
,
Greenwood
CM
, et al.
Toward precision medicine: TBC1D4 disruption is common among the inuit and leads to underdiagnosis of Type 2 diabetes
.
Diabetes Care
.
2016
Nov
;
39
(
11
):
1889
95
.
[PubMed]
0149-5992
18.
Holstein
A
,
Plaschke
A
,
Ptak
M
,
Egberts
EH
,
El-Din
J
,
Brockmöller
J
, et al.
Association between CYP2C9 slow metabolizer genotypes and severe hypoglycaemia on medication with sulphonylurea hypoglycaemic agents
.
Br J Clin Pharmacol
.
2005
Jul
;
60
(
1
):
103
6
.
[PubMed]
0306-5251
19.
Ragia
G
,
Petridis
I
,
Tavridou
A
,
Christakidis
D
,
Manolopoulos
VG
.
Presence of CYP2C9*3 allele increases risk for hypoglycemia in Type 2 diabetic patients treated with sulfonylureas
.
Pharmacogenomics
.
2009
Nov
;
10
(
11
):
1781
7
.
[PubMed]
1462-2416
20.
Niemi
M
,
Leathart
JB
,
Neuvonen
M
,
Backman
JT
,
Daly
AK
,
Neuvonen
PJ
.
Polymorphism in CYP2C8 is associated with reduced plasma concentrations of repaglinide
.
Clin Pharmacol Ther
.
2003
Oct
;
74
(
4
):
380
7
.
[PubMed]
0009-9236
21.
Nies
AT
,
Koepsell
H
,
Winter
S
,
Burk
O
,
Klein
K
,
Kerb
R
, et al.
Expression of organic cation transporters OCT1 (SLC22A1) and OCT3 (SLC22A3) is affected by genetic factors and cholestasis in human liver
.
Hepatology
.
2009
Oct
;
50
(
4
):
1227
40
.
[PubMed]
0270-9139
22.
Shu
Y
,
Sheardown
SA
,
Brown
C
,
Owen
RP
,
Zhang
S
,
Castro
RA
, et al.
Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action
.
J Clin Invest
.
2007
May
;
117
(
5
):
1422
31
.
[PubMed]
0021-9738
23.
Pawlyk
AC
,
Giacomini
KM
,
McKeon
C
,
Shuldiner
AR
,
Florez
JC
.
Metformin pharmacogenomics: current status and future directions
.
Diabetes
.
2014
Aug
;
63
(
8
):
2590
9
.
[PubMed]
0012-1797
24.
Mahrooz
A
,
Parsanasab
H
,
Hashemi-Soteh
MB
,
Kashi
Z
,
Bahar
A
,
Alizadeh
A
, et al.
The role of clinical response to metformin in patients newly diagnosed with type 2 diabetes: a monotherapy study
.
Clin Exp Med
.
2015
May
;
15
(
2
):
159
65
.
[PubMed]
1591-8890
25.
Zhou
K
,
Bellenguez
C
,
Spencer
CC
,
Bennett
AJ
,
Coleman
RL
,
Tavendale
R
, et al.;
GoDARTS and UKPDS Diabetes Pharmacogenetics Study Group
;
Wellcome Trust Case Control Consortium 2
;
MAGIC investigators
.
Common variants near ATM are associated with glycemic response to metformin in type 2 diabetes
.
Nat Genet
.
2011
Feb
;
43
(
2
):
117
20
.
[PubMed]
1061-4036
26.
Chen
M
,
Hu
C
,
Zhang
R
,
Jiang
F
,
Wang
J
,
Peng
D
, et al.
Association of PAX4 genetic variants with oral antidiabetic drugs efficacy in Chinese type 2 diabetes patients
.
Pharmacogenomics J
.
2014
Oct
;
14
(
5
):
488
92
.
[PubMed]
1470-269X
27.
Chen
M
,
Hu
C
,
Zhang
R
,
Jiang
F
,
Wang
J
,
Peng
D
, et al.
A variant of PSMD6 is associated with the therapeutic efficacy of oral antidiabetic drugs in Chinese type 2 diabetes patients
.
Sci Rep
.
2015
May
;
5
(
1
):
10701
.
[PubMed]
2045-2322
28.
Dujic
T
,
Zhou
K
,
Donnelly
LA
,
Tavendale
R
,
Palmer
CN
,
Pearson
ER
.
Association of organic cation transporter 1 with intolerance to metformin in type 2 diabetes: A GoDARTS Study
.
Diabetes
.
2015
May
;
64
(
5
):
1786
93
.
[PubMed]
0012-1797
29.
Zhou
K
,
Donnelly
LA
,
Kimber
CH
,
Donnan
PT
,
Doney
AS
,
Leese
G
, et al.
Reduced-function SLC22A1 polymorphisms encoding organic cation transporter 1 and glycemic response to metformin: a GoDARTS study
.
Diabetes
.
2009
Jun
;
58
(
6
):
1434
9
.
[PubMed]
0012-1797
30.
Dawed
AY
,
Donnelly
L
,
Tavendale
R
,
Carr
F
,
Leese
G
,
Palmer
CN
, et al.
CYP2C8 and SLCO1B1 Variants and therapeutic response to thiazolidinediones in patients with type 2 diabetes
.
Diabetes Care
.
2016
Nov
;
39
(
11
):
1902
8
.
[PubMed]
0149-5992
31.
Aghaei Meybodi
HR
,
Hasanzad
M
,
Larijani
B
.
Path to personalized medicine for type 2 diabetes mellitus: reality and Hope
.
Acta Med Iran
.
2017
Mar
;
55
(
3
):
166
74
.
[PubMed]
1735-9694
32.
Zimdahl
H
,
Ittrich
C
,
Graefe-Mody
U
,
Boehm
BO
,
Mark
M
,
Woerle
HJ
, et al.
Influence of TCF7L2 gene variants on the therapeutic response to the dipeptidylpeptidase-4 inhibitor linagliptin
.
Diabetologia
.
2014
Sep
;
57
(
9
):
1869
75
.
[PubMed]
0012-186X
33.
Prasad
RB
,
Groop
L
.
Genetics of type 2 diabetes-pitfalls and possibilities
.
Genes (Basel)
.
2015
Mar
;
6
(
1
):
87
123
.
[PubMed]
2073-4425
34.
’t Hart
LM
,
Fritsche
A
,
Nijpels
G
,
van Leeuwen
N
,
Donnelly
LA
,
Dekker
JM
, et al.
The CTRB1/2 locus affects diabetes susceptibility and treatment via the incretin pathway
.
Diabetes
.
2013
Sep
;
62
(
9
):
3275
81
.
[PubMed]
0012-1797
35.
Dawed
AY
,
Zhou
K
,
Pearson
ER
.
Pharmacogenetics in type 2 diabetes: influence on response to oral hypoglycemic agents
.
Pharmgenomics Pers Med
.
2016
Apr
;
9
:
17
29
.
[PubMed]
1178-7066
36.
Mohan
V
,
Yang
W
,
Son
HY
,
Xu
L
,
Noble
L
,
Langdon
RB
, et al.
Efficacy and safety of sitagliptin in the treatment of patients with type 2 diabetes in China, India, and Korea
.
Diabetes Res Clin Pract
.
2009
Jan
;
83
(
1
):
106
16
.
[PubMed]
0168-8227
37.
Sudhakaran
C
,
Kishore
U
,
Anjana
RM
,
Unnikrishnan
R
,
Mohan
V
.
Effectiveness of sitagliptin in asian Indian patients with type 2 diabetes-an Indian tertiary diabetes care center experience
.
Diabetes Technol Ther
.
2011
Jan
;
13
(
1
):
27
32
.
[PubMed]
1520-9156
38.
Kim
YG
,
Hahn
S
,
Oh
TJ
,
Kwak
SH
,
Park
KS
,
Cho
YM
.
Differences in the glucose-lowering efficacy of dipeptidyl peptidase-4 inhibitors between Asians and non-Asians: a systematic review and meta-analysis
.
Diabetologia
.
2013
Apr
;
56
(
4
):
696
708
.
[PubMed]
0012-186X
39.
Park
H
,
Park
C
,
Kim
Y
,
Rascati
KL
.
Efficacy and safety of dipeptidyl peptidase-4 inhibitors in type 2 diabetes: meta-analysis
.
Ann Pharmacother
.
2012
Nov
;
46
(
11
):
1453
69
.
[PubMed]
1060-0280
40.
Kim
YG
,
Hahn
S
,
Oh
TJ
,
Park
KS
,
Cho
YM
.
Differences in the HbA1c-lowering efficacy of glucagon-like peptide-1 analogues between Asians and non-Asians: a systematic review and meta-analysis
.
Diabetes Obes Metab
.
2014
Oct
;
16
(
10
):
900
9
.
[PubMed]
1462-8902
41.
Staimez
LR
,
Weber
MB
,
Ranjani
H
,
Ali
MK
,
Echouffo-Tcheugui
JB
,
Phillips
LS
, et al.
Evidence of reduced β-cell function in Asian Indians with mild dysglycemia
.
Diabetes Care
.
2013
Sep
;
36
(
9
):
2772
8
.
[PubMed]
0149-5992
42.
Narayan
KM
.
Type 2 Diabetes: why we are winning the battle but losing the war? 2015 Kelly West Award Lecture
.
Diabetes Care
.
2016
May
;
39
(
5
):
653
63
.
[PubMed]
0149-5992
43.
Schwartz
SS
,
Epstein
S
,
Corkey
BE
,
Grant
SF
,
Gavin
JR
 3rd
,
Aguilar
RB
.
The time is right for a new classification system for diabetes: rationale and implications of the β-cell-centric classification schema
.
Diabetes Care
.
2016
Feb
;
39
(
2
):
179
86
.
[PubMed]
0149-5992
44.
Schwartz
SS
,
Epstein
S
,
Corkey
BE
,
Grant
SF
,
Gavin Iii
JR
,
Aguilar
RB
, et al.
A unified pathophysiologic construct of diabetes and its complications
.
Trends Endocrinol Metab
.
2017
Sep
;
28
(
9
):
645
55
.
[PubMed]
1043-2760
45.
Hattersley
AT
,
Patel
KA
.
Precision diabetes: learning from monogenic diabetes
.
Diabetologia
.
2017
May
;
60
(
5
):
769
77
.
[PubMed]
0012-186X
46.
Pearson
ER
,
Flechtner
I
,
Njølstad
PR
,
Malecki
MT
,
Flanagan
SE
,
Larkin
B
, et al.;
Neonatal Diabetes International Collaborative Group
.
Switching from insulin to oral sulfonylureas in patients with diabetes due to Kir6.2 mutations
.
N Engl J Med
.
2006
Aug
;
355
(
5
):
467
77
.
[PubMed]
0028-4793
47.
Sagen
JV
,
Raeder
H
,
Hathout
E
,
Shehadeh
N
,
Gudmundsson
K
,
Baevre
H
, et al.
Permanent neonatal diabetes due to mutations in KCNJ11 encoding Kir6.2: patient characteristics and initial response to sulfonylurea therapy
.
Diabetes
.
2004
Oct
;
53
(
10
):
2713
8
.
[PubMed]
0012-1797
48.
Beltrand
J
,
Elie
C
,
Busiah
K
,
Fournier
E
,
Boddaert
N
,
Bahi-Buisson
N
, et al.;
GlidKir Study Group
.
Sulfonylurea therapy benefits neurological and psychomotor functions in patients with neonatal diabetes owing to potassium channel mutations
.
Diabetes Care
.
2015
Nov
;
38
(
11
):
2033
41
.
[PubMed]
0149-5992
49.
Gloyn
AL
,
Diatloff-Zito
C
,
Edghill
EL
,
Bellanné-Chantelot
C
,
Nivot
S
,
Coutant
R
, et al.
KCNJ11 activating mutations are associated with developmental delay, epilepsy and neonatal diabetes syndrome and other neurological features
.
Eur J Hum Genet
.
2006
Jul
;
14
(
7
):
824
30
.
[PubMed]
1018-4813
50.
Jahnavi
S
,
Poovazhagi
V
,
Mohan
V
,
Bodhini
D
,
Raghupathy
P
,
Amutha
A
, et al.
Clinical and molecular characterization of neonatal diabetes and monogenic syndromic diabetes in Asian Indian children
.
Clin Genet
.
2013
May
;
83
(
5
):
439
45
.
[PubMed]
0009-9163
51.
Hattersley
AT
,
Ashcroft
FM
.
Activating mutations in Kir6.2 and neonatal diabetes: new clinical syndromes, new scientific insights, and new therapy
.
Diabetes
.
2005
Sep
;
54
(
9
):
2503
13
.
[PubMed]
0012-1797
52.
Radha
V
,
Ramya
B
,
Gopi
S
,
Kavitha
B
,
Preetika
S
,
Thai
K
, et al.
Successful transition to sulphonylurea therapy from insulin in a child with permanent neonatal diabetes due to a KCNJ11 gene mutation
.
J Diabetol
.
2018
;
9
(
2
):
65
7
.
53.
De Franco
E
,
Flanagan
SE
,
Houghton
JA
,
Lango Allen
H
,
Mackay
DJ
,
Temple
IK
, et al.
The effect of early, comprehensive genomic testing on clinical care in neonatal diabetes: an international cohort study
.
Lancet
.
2015
Sep
;
386
(
9997
):
957
63
.
[PubMed]
0140-6736
54.
Villamayor
L
,
Rodríguez-Seguel
E
,
Araujo
R
,
Carrasco
M
,
Bru-Tarí
E
,
Mellado-Gil
JM
, et al.
GATA6 Controls Insulin Biosynthesis and Secretion in Adult β-Cells
.
Diabetes
.
2018
Mar
;
67
(
3
):
448
60
.
[PubMed]
0012-1797
55.
Cavé
H
,
Polak
M
,
Drunat
S
,
Denamur
E
,
Czernichow
P
.
Refinement of the 6q chromosomal region implicated in transient neonatal diabetes
.
Diabetes
.
2000
Jan
;
49
(
1
):
108
13
.
[PubMed]
0012-1797
56.
Rubio-Cabezas
O
,
Flanagan
SE
,
Damhuis
A
,
Hattersley
AT
,
Ellard
S
.
KATP channel mutations in infants with permanent diabetes diagnosed after 6 months of life
.
Pediatr Diabetes
.
2012
Jun
;
13
(
4
):
322
5
.
[PubMed]
1399-543X
57.
Jahnavi
S
,
Poovazhagi
V
,
Kanthimathi
S
,
Balamurugan
K
,
Bodhini
D
,
Yadav
J
, et al.
Novel ABCC8 (SUR1) gene mutations in Asian Indian children with congenital hyperinsulinemic hypoglycemia
.
Ann Hum Genet
.
2014
Sep
;
78
(
5
):
311
9
.
[PubMed]
0003-4800
58.
Olsen
BS
,
Hahnemann
JM
,
Schwartz
M
,
Østergaard
E
.
Thiamine-responsive megaloblastic anaemia: a cause of syndromic diabetes in childhood
.
Pediatr Diabetes
.
2007
Aug
;
8
(
4
):
239
41
.
[PubMed]
1399-543X
59.
Bacchetta
R
,
Barzaghi
F
,
Roncarolo
MG
.
From IPEX syndrome to FOXP3 mutation: a lesson on immune dysregulation
.
Ann N Y Acad Sci
.
2018
Apr
;
1417
(
1
):
5
22
.
[PubMed]
0077-8923
60.
Tattersall
RB
,
Fajans
SS
,
Arbor
A
.
A difference between the inheritance of classical juvenile-onset and maturity-onset type diabetes of young people
.
Diabetes
.
1975
Jan
;
24
(
1
):
44
53
.
[PubMed]
0012-1797
61.
Anuradha
S
,
Radha
V
,
Mohan
V
.
Association of novel variants in the hepatocyte nuclear factor 4A gene with maturity onset diabetes of the young and early onset type 2 diabetes
.
Clin Genet
.
2011
Dec
;
80
(
6
):
541
9
.
[PubMed]
0009-9163
62.
Anuradha
S
,
Radha
V
,
Deepa
R
,
Hansen
T
,
Carstensen
B
,
Pedersen
O
, et al.
A prevalent amino acid polymorphism at codon 98 (Ala98Val) of the hepatocyte nuclear factor-1alpha is associated with maturity-onset diabetes of the young and younger age at onset of type 2 diabetes in Asian Indians
.
Diabetes Care
.
2005
Oct
;
28
(
10
):
2430
5
.
[PubMed]
0149-5992
63.
Murphy
R
,
Ellard
S
,
Hattersley
AT
.
Clinical implications of a molecular genetic classification of monogenic beta-cell diabetes
.
Nat Clin Pract Endocrinol Metab
.
2008
Apr
;
4
(
4
):
200
13
.
[PubMed]
1745-8366
64.
Jesić
MD
,
Sajić
S
,
Jesić
MM
,
Maringa
M
,
Micić
D
,
Necić
S
.
A case of new mutation in maturity-onset diabetes of the young type 3 (MODY 3) responsive to a low dose of sulphonylurea
.
Diabetes Res Clin Pract
.
2008
Jul
;
81
(
1
):
e1
3
.
[PubMed]
0168-8227
65.
Pearson
ER
,
Badman
MK
,
Lockwood
CR
,
Clark
PM
,
Ellard
S
,
Bingham
C
, et al.
Contrasting diabetes phenotypes associated with hepatocyte nuclear factor-1alpha and -1beta mutations
.
Diabetes Care
.
2004
May
;
27
(
5
):
1102
7
.
[PubMed]
0149-5992
66.
Jahnavi
S
,
Poovazhagi
V
,
Mohan
V
,
Radha
V
.
Neonatal diabetes and hyperinsulinemia: the Indian experience
.
J Neonatol
.
2013
;
27
:
15
23
.
67.
Varadarajan
P
,
Ananthanarayanan
K
,
Mirna
K
,
Suresh
J
,
Venkatesan
R
,
Mohan
V
.
Clinical profile and outcome of persistent hyperinsulinemic hypoglycemia of infancy
.
Pediatr Oncall
.
2013
;
50
:
759
63
.0973-0966
68.
Pearson
ER
,
Starkey
BJ
,
Powell
RJ
,
Gribble
FM
,
Clark
PM
,
Hattersley
AT
.
Genetic cause of hyperglycaemia and response to treatment in diabetes
.
Lancet
.
2003
Oct
;
362
(
9392
):
1275
81
.
[PubMed]
0140-6736
69.
Pearson
ER
,
Liddell
WG
,
Shepherd
M
,
Corrall
RJ
,
Hattersley
AT
.
Sensitivity to sulphonylureas in patients with hepatocyte nuclear factor-1alpha gene mutations: evidence for pharmacogenetics in diabetes
.
Diabet Med
.
2000
Jul
;
17
(
7
):
543
5
.
[PubMed]
0742-3071
70.
Pearson
ER
,
Pruhova
S
,
Tack
CJ
,
Johansen
A
,
Castleden
HA
,
Lumb
PJ
, et al.
Molecular genetics and phenotypic characteristics of MODY caused by hepatocyte nuclear factor 4alpha mutations in a large European collection
.
Diabetologia
.
2005
May
;
48
(
5
):
878
85
.
[PubMed]
0012-186X
71.
Chakera
AJ
,
Steele
AM
,
Gloyn
AL
,
Shepherd
MH
,
Shields
B
,
Ellard
S
, et al.
Recognition and management of individuals with hyperglycemia because of a heterozygous glucokinase mutation
.
Diabetes Care
.
2015
Jul
;
38
(
7
):
1383
92
.
[PubMed]
0149-5992
72.
Kanthimathi
S
,
Jahnavi
S
,
Balamurugan
K
,
Ranjani
H
,
Sonya
J
,
Goswami
S
, et al.
Glucokinase gene mutations (MODY 2) in Asian Indians
.
Diabetes Technol Ther
.
2014
Mar
;
16
(
3
):
180
5
.
[PubMed]
1520-9156
73.
Spyer
G
,
Macleod
KM
,
Shepherd
M
,
Ellard
S
,
Hattersley
AT
.
Pregnancy outcome in patients with raised blood glucose due to a heterozygous glucokinase gene mutation
.
Diabet Med
.
2009
Jan
;
26
(
1
):
14
8
.
[PubMed]
0742-3071
74.
Radha
V
,
Ek
J
,
Anuradha
S
,
Hansen
T
,
Pedersen
O
,
Mohan
V
.
Identification of novel variants in the hepatocyte nuclear factor-1alpha gene in South Indian patients with maturity onset diabetes of young
.
J Clin Endocrinol Metab
.
2009
Jun
;
94
(
6
):
1959
65
.
[PubMed]
0021-972X
75.
Balamurugan
K
,
Bjørkhaug
L
,
Mahajan
S
,
Kanthimathi
S
,
Njølstad
PR
,
Srinivasan
N
, et al.
Structure-function studies of HNF1A (MODY3) gene mutations in South Indian patients with monogenic diabetes
.
Clin Genet
.
2016
Dec
;
90
(
6
):
486
95
.
[PubMed]
0009-9163
76.
Kanthimathi
S
,
Balamurugan
K
,
Shanthi Rani
CS
,
Mohan
V
,
Radha
V
.
Identification and functional characterization of hepatocyte nuclear factor -1b (MODY 5) gene mutations in Indian diabetic patients with renal abnormalities
.
Ann Hum Genet
.
2015
;
79
:
10
9
.
[PubMed]
0003-4800
77.
Mohan
V
,
Radha
V
,
Nguyen
TT
,
Stawiski
EW
,
Pahuja
KB
,
Goldstein
LD
, et al.
Comprehensive genomic analysis identifies pathogenic variants in maturity-onset diabetes of the young (MODY) patients in South India. BMC Med Genet.
2018
13;19:22.
Open Access License / Drug Dosage / Disclaimer
This article is licensed under the Creative Commons Attribution-NonCommercial 4.0 International License (CC BY-NC). Usage and distribution for commercial purposes requires written permission. Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug. Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.