You are on page 1of 19

REVIEW OF THERAPEUTICS

Using Personalized Medicine in the Management of


Diabetes Mellitus

Nina Elk1* and Otito F. Iwuchukwu2


1
Division of Pharmacy Practice, Fairleigh Dickinson University School of Pharmacy, Florham Park, New Jersey;
2
Division of Pharmaceutical Sciences, Fairleigh Dickinson University School of Pharmacy, Florham Park, New
Jersey

Diabetes mellitus is a worldwide problem with an immense pharmacoeconomic burden. The multifac-
torial and complex nature of the disease lends itself to personalized pharmacotherapeutic approaches
to treatment. Variability in individual risk and subsequent development of diabetes has been reported
in addition to differences in response to the many oral glucose lowering therapies currently available
for diabetes pharmacotherapy. Pharmacogenomic studies have attempted to uncover the heritable com-
ponents of individual variability in risk susceptibility and response to pharmacotherapy. We review
the current pharmacogenomics evidence as it relates to common oral glucose lowering therapies and
how they can be utilized in the management of polygenic and monogenic forms of diabetes. Evidence
supports the use of genetic testing and personalized approaches to the treatment of monogenic dia-
betes of the young. The data are not as robust for the current application of pharmacogenetic
approaches to the treatment of polygenic type 2 diabetes mellitus, but there are suggestions as to
future applications in this regard. We reviewed pertinent primary literature sources as well as current
evidence-based guidelines on diabetes management.
KEY WORDS pharmacogenomics, diabetes, monogenic diabetes syndrome, maturity-onset diabetes of
the young, oral glucose lowering agents.
(Pharmacotherapy 2017;37(9):1131–1149) doi: 10.1002/phar.1976

Diabetes mellitus (DM) presents a significant Global Report, the number of people diagnosed
public health problem worldwide and is recog- with DM worldwide has been increasing, reaching
nized as a cause of premature death and disabil- 422 million adults in 2014 and causing 1.5 million
ity.1 According to the World Health Organization’s deaths.2 In the United States, although newly diag-
nosed cases of DM are declining, DM remains at
an all-time high with 29 million diagnoses
The authors, employees of Fairleigh Dickinson Univer- reported for 2016 (https://www.cdc.gov/chronicdis
sity School of Pharmacy, certify that they have no affilia-
tions with or involvement in any organization or entity
ease/resources/publications/aag/diabetes.htm). DM
with any financial interest (such as honoraria; educational was the seventh leading cause of death in 2013,
grants; participation in speakers’ bureaus; membership, and data indicate that patients with diabetes are
employment, consultancies, stock ownership, or other twice as likely to develop cardiac complications at
equity interest; and expert testimony or patent-licensing a younger age compared with people who do not
arrangements), or nonfinancial interest (such as personal or
professional relationships, affiliations, knowledge or beliefs)
have the disease.3 The economic burden of DM
in the subject matter or materials discussed in this manu- remains high. Direct and indirect costs stand at
script. $245 billion and cost of medical expenditures for
*Address for correspondence: Nina Elk, School of Phar- individuals with DM at $17,000 annually, which is
macy, Fairleigh Dickinson University, 230 Park Avenue, reportedly 2.3 times greater than for those without
M-SP1-01, Florham Park, NJ 07932; e-mail: NinaElk@fdu.edu.
Ó 2017 Pharmacotherapy Publications, Inc.
DM.4
1132 PHARMACOTHERAPY Volume 37, Number 9, 2017

Prevention of DM, early diagnosis, and delay Type 2 Diabetes Mellitus: A Polygenic,
of disease progression are critical measures to Multifactorial Disease
avoid life-threatening complications. Current
guidelines emphasize the importance of personal- T2DM is the most prevalent type of diabetes,
ized care for patients with DM and recommend and its polygenicity is supported by many lines
use of patient-specific pharmacotherapy of evidence showing both a distinct familial
approaches to treatment.5 An algorithmic aggregation and a lack of Mendelian segrega-
approach to the management of DM incorporates tion.6 Pathophysiology of T2DM involves multi-
steps such as identification of diabetes type, gly- ple abnormalities in the regulation of glucose
cemic target selection, use of medications based homeostasis (Figure 2). The search for suscepti-
on proven efficacy, and side-effect profiles.5 bility risk genes involved in the pathophysiology
In addition, addressing comorbidities attributing of T2DM and its observed phenotypes has been
to cardiovascular diseases and counseling ongoing for decades. Several strategies that have
patients on a healthy lifestyle are components of been used include linkage, candidate gene, gen-
personalized care for diabetes management. ome, and exome-wide association studies.9
Genetic findings in DM risk assessment, patho- Newer approaches, such as the reverse genetics
physiology, disposition, and efficacy of drugs phenome-wide approach of surveying clinically
used in DM allow expanding the definition of validated phenotypes with variants in known
patient-specific therapy (Figure 1). This article susceptibility genes, have also been used to vali-
reviews the genetic findings in pathophysiology date diabetes-linked genes and pathways.10
and risk association of DM and pharmacogenet- Candidate gene studies (CGS) focus on genes
ics of common oral glucose lowering agents, fol- related to the disease in question and a priori
lowed by a discussion of a pharmacogenomic knowledge of any mechanistic roles in the dis-
contribution to personalized therapy. ease pathway. Genes involved in glucose home-
ostasis, specifically those encoding pathways of
insulin production, secretion, and response
Pharmacogenomic Landscape of Diabetes (such as the insulin receptor [INSR], glycogen
Mellitus synthase 1 [GYS1], glucose transporter 4
[GLUT4], and insulin receptor site-1 [IRS-1])
Although DM is classified into four general
were the main focus of T2DM CGS.11, 12 Overall,
categories based on pathophysiology, further
CGS contribution to valid genetic associations in
differentiation based on genetic components is
T2DM was limited and sometimes not repro-
possible (Table 1). For the remainder of this
ducible due to the pleiotropic nature of the dis-
section, we focus on the polygenic type 2 DM
ease, small sample sizes, high genotyping costs,
(T2DM) with some discussion on the monogenic
and low genetic signal throughput. Despite these
forms of diabetes that include maturity-onset
shortcomings, CGS led to the identification of
diabetes of the young (MODY) and neonatal dia-
common variants in confirmed susceptibility
betes mellitus (NDM).
genes such as peroxisome proliferator-activated
receptor gamma (PPAR-c) and KCNJ11 encoding
the inwardly rectifying Kir6.2 component of the
b-cell adenosine triphosphate (ATP)-sensitive
potassium channel (KATP channel).13
Genome-wide association studies (GWAS) are
comprehensive studies that use an unbiased and
hypothesis-free approach to interrogate the
whole genome for associations between genetic
variants and observed phenotypes.7 Because
GWAS are a special type of case-control studies
where individual genomes of subjects with dis-
ease (cases) are scanned for genetic markers dif-
fering in frequency from control subjects, there
is potential power to discover novel genetic loci
underlying predisposition to and development of
T2DM.7 Some limitations (small sample size,
Figure 1. Factors to consider when designing diabetes
mellitus (DM) treatment regimen.
low throughput) of diabetes CGS were overcome
PERSONALIZED MEDICINE IN DIABETES MANAGEMENT Elk and Iwuchukwu 1133
Table 1. Differentiation of Diabetes Mellitus Types5–8
DM type Prevalence Pathophysiology Genetic component
Type 1 5–10% Autoantibodies to pancreatic Mostly associated with genetic
islet cells and insulin are variation of HLA association
present that cause pancreatic genes responsible for immune
b-cell destruction leading to system response and pancreatic
insulin deficiency. b-cell function. Other genes are
linked to type 1 DM.a
Type 2 90–95% Increase in glucose production Mutations in specific genes
by the liver, insulin resistance associated with disorders
in target tissues, and dysfunction of glucose homeostasis.
of pancreatic b cells in the
pancreas causing impairment
of insulin secretion.
Monogenic diabetes <5% Insulin is produced but not Mutations in a gene that
syndrome: NDM secreted through KATP channels encodes KATP channels in
in pancreatic b cells. pancreatic b cells.
Monogenic diabetes <5% In MODY 1 and MODY 3, Mutations in the gene that
syndrome: MODY hyperglycemia is present due encodes HNF4a in MODY 1
to partial pancreatic b-cell dysfunction. and HNF1a in MODY 3
In MODY 2, hyperglycemia Mutations in the gene that
is due to dysfunction of encodes GCK and several
glucose-level monitoring by transcription factors relevant
pancreatic b cells. to pancreatic b-cell development.
DM = diabetes mellitus; GCK = glucokinase; HLA = human leukocyte antigen; HNF = hepatocyte nuclear factor; KATP channel = ATP-sensi-
tive K+ channel; MODY = maturity-onset diabetes of the young; NDM = neonatal diabetes mellitus.
a
For a complete list of genes associated with type 1 DM, visit https://ghr.nlm.nih.gov/condition/type-1-diabetes#.

with GWAS, and due to the high-powered nat- only a fraction of heritability was attributable to
ure of this approach, many genetic loci were common variants and that combinations of rare
both discovered and/or validated.11, 14 The initial alleles of small effect size were more likely to
set of GWAS studies on diabetes was published explain a greater fraction of T2DM heritability.19
a decade ago, and about a dozen loci were This rare variant hypothesis is still unproven. A
strongly associated with T2DM risk across these very recent study by two of the largest diabetes
studies.15 Indeed, the strongest genetic signal(s) GWAS consortia concluded that large-scale
associated to date with diabetes susceptibility sequencing did not support the idea of a major
was found with single nucleotide polymorphisms role of lower frequency rare variants in predis-
(SNPs) in the transcription factor 7-like 2 position to T2DM.17
(TCF7L2) gene. This loci was discovered by
whole-genome microsatellite marker analysis in
Monogenic Diabetes Syndrome
a cohort of ~1000 Icelandic subjects16 and sub-
sequently validated and replicated in T2DM The monogenic diabetes syndrome covers a
GWAS.14, 17 The protein encoded by TCF7L2 is heterogeneous group of single-gene autosomally
a high-mobility group box containing transcrip- inherited diabetes that cannot be classified as
tion factor postulated to affect diabetes risk type 1 or type 2 DM. The syndrome can be fur-
through pancreatic b-cell dysfunction via the ther divided into two main forms, MODY and
Wnt signaling pathway.16 Other validated signals NDM.
reported include SNPs in the KCNJ11, PPAR-c,
Fat Mass and Obesity (FTO) associated genes15
Maturity-Onset Diabetes of the Young
(Table 2). At the time of last access, the publicly
available NHGRI-EBI Catalog of published MODY (unlike polygenic T2DM) arises from
GWAS18 lists over 157 studies on diabetes with monogenic defects in pancreatic b-cell function
more than 1500 associations at genome-wide with little or no defect in insulin action. The clini-
significance levels (p=5 9 108). One limiting cal features of MODY include autosomal domi-
assumption of the GWAS approach is that nant inheritance, early onset (before age 25),
genetic variants conferring disease susceptibility absence of obesity, and evidence of b-cell function
are common (more than 5% frequency) in the as seen with endogenous insulin secretion.20 Cur-
studied populations,14 an assumption that has rently ~13 different types of MODY (1–14) exist,
not held true with T2DM. It was postulated that associated with abnormalities in more than six
1134 PHARMACOTHERAPY Volume 37, Number 9, 2017

Figure 2. Schematic presentation of type 2 diabetes mellitus (T2DM) pathophysiology. Pathophysiology of T2DM involves
deficiency of insulin action that leads to hyperglycemia and inability to maintain glucose homeostasis. Deficiency of insulin
action may be due to decrease in amount of insulin secreted by islet cells of the pancreas, insulin resistance with skeletal
muscles’ inability to uptake glucose for utilization, or a combination of both. Decreased insulin secretion results from b-cell
dysfunction, decrease in b-cell mass, and reduced action of glucagon-like peptide 1 hormone that is involved in insulin
secretion. Insulin resistance linked to obesity is due to a decreased number of insulin receptors, increased fatty acids, and
triglycerides in adipose tissue interfering with insulin action, as well as causing an increase in production of glucose by the
liver. Disease progresses with unresolved hyperglycemia, increase in gluconeogenesis, and loss of b-cell function.

different loci on different chromosomes and sub- The resultant impaired hepatic cell glucose
classified by the deficient gene causing the phosphorylation is responsible for the very mild
observable phenotype.21 MODY makes up ~1–4% hyperglycemia observed in patients with
of all pediatric DM cases, and because presenting MODY 28, 22 (Table 3).
characteristics include those associated with
either T1DM (early onset, lean body mass) or
Neonatal Diabetes Mellitus Syndrome
T2DM (preserved b-cell function, familial aggre-
gation), misdiagnosis is common.22, 23 NDM syndrome is a rare form of diabetes pre-
MODY 1, 2, and 3 are the most well character- senting within the first 6 months of life. Infants
ized of all MODY subtypes. MODY 1 makes up with NDM do not produce enough insulin, lead-
~10% of cases in characterized cohorts of MODY ing to increased blood glucose levels. In about
subjects.21 It is caused by mutations in HNF4a, half of presenting patients, the condition persists
encoding the transcription factor hepatocyte and is termed permanent neonatal diabetes mel-
nuclear factor 4-a.22 A defect in a similar modi- litus (PNDM). In the other half of cases, the
fier gene, HNF1a, is responsible for the most condition disappears during infancy and is ter-
common subtype MODY 3 that together with med transient neonatal diabetes mellitus
MODY 2 makes up ~50% of all cases.21 These (TNDM). NDM has a number of genetic causes
hepatic transcription factors promote the tran- attributed to it including variations in genes
scription of multiple genes related to glucose such as KCNJ11, ABCC8, GCK, INS, and
metabolism and insulin production and secretion. ZFP57.23–26 The most common causes are muta-
MODY 2 is due to mutations in the glucoki- tions in KCNJ11 and ABCC8, the two genes
nase (GCK) gene, causing decreased function of encoding different subunits of the ATP-depen-
glucokinase, the enzyme responsible for blood dent potassium channel in pancreatic islet b
glucose level homeostasis in pancreatic b cells.20 cells. These mutations serve to decrease insulin
PERSONALIZED MEDICINE IN DIABETES MANAGEMENT Elk and Iwuchukwu 1135
Table 2. Selected Genes and Single Nucleotide Polymorphisms Associated with Insulin Action or b-Cell Function in
T2DM Risk/Susceptibility GWAS
Overall Postulated
Gene Chromosome rs number odds ratio Function mechanism Year
ADAMTS911, 14
3 rs4607103 1.09 Secreted Insulin action 2008
metalloproteinase
ADCY514 3 rs11708067 1.12 Adenylyl cyclase Insulin action 2010
BCAR114, 17
16 rs7202877 1.12 Docking protein Regulation of 2012
b-cell function
BCL11A14, 17
2 rs243021 1.08 Zinc finger protein Regulation of 2010
b-cell function
CCND217 12 rs76895963 0.53 Cyclin D2 Cell cycle 2014
regulator/Enhanced
insulin secretion
CDKAL19, 11, 14, 17
6 rs7754840 1.15 Methylthiotransferase Regulation of 2007
b-cell function
CDKN2A/B9, 11, 14, 17
9 rs7754840 1.20 Cyclin-dependent Regulation of 2007
kinase inhibitor b-cell function
FTO9, 11, 14, 17
16 rs8050136 1.27 Fat Mass and Obesity Insulin action 2007
Associated/Nucleic
acid demethylase
GCKR11, 17
2 rs780094 1.08 Glucokinase Insulin action 2007
regulator protein
HHEX/IDE 9, 11, 14, 17
10 rs1111875 1.15 Transcriptional repressor Intracellular 2007
insulin degradation
IGF2BP29, 11, 14, 17
3 rs4402960 1.17 Insulinlike growth factor Regulation of 2007
II mRNA-binding protein b-cell function
IRS111, 14, 17 2 rs2943640 1.12 Docking/binding protein Insulin action 2009
JAZF19, 11, 14, 17
7 rs864745 1.10 Zinc finger protein Regulation of 2008
b-cell function
KCNJ1111, 17
11 rs5219 1.14 Inwardly rectifying Regulation of 2003
potassium channel Kir6.2 insulin secretion
KCNQ1 9, 11, 14, 17 11 rs2237892 1.23 Potassium channel b-cell function 2008
NOTCH2 9, 11, 14, 17 1 rs10923931 1.13 Transmembrane receptor Pancreatic 2008
cell development
9, 11, 14, 15, 17
PPAR-c 3 rs1801282 1.11 Peroxisome Regulation 2000
proliferator-activated of insulin action
receptor
SLC30A8 9, 11, 14, 17
8 rs13266634 1.15 b-cell zinc efflux transporter Insulin storage 2007
and secretion
TCF7L214, 15
10 rs7903146 1.4 T-cell transcription factor b-cell function 2006
TSPAN8 9, 11, 14, 17
12 rs7961581 1.09 Cell surface glycoprotein b-cell function 2008
GWAS = genome-wide association study; rs = reference sequence. Note: The rs numbers indicate the reference single nucleotide polymor-
phism (SNP) cluster ID used for identification within the SNP public archive database (https://www.ncbi.nlm.nih.gov/projects/SNP/). All
GWAS are documented in the Catalog of Published Genome-Wide Association Studies (http://www.ebi.ac.uk/gwas).

secretion by preventing membrane depolariza- Metformin


tion (Table 3).
Metformin belongs to the biguanide class of
oral glucose lowering therapies (OGLTs).
Pharmacogenetic-Based Associations for Although many aspects of metformin’s action are
Common Oral Glucose Lowering Therapies still not well established, it has been postulated
to exert its glucose-lowering effects by many
Various agents are available for the treatment pleiotropic mechanisms.28 One major hypothesis
of DM. They differ in their mechanisms of action is metformin-induced activation of AMP-acti-
and efficacy, and ability to attain DM treatment vated protein kinase (AMPK).29 In addition,
goals such as prevention of macrovascular and metformin prevents hyperglycemia by decreasing
microvascular complications. In addition, antihy- intestinal glucose absorption and increasing
perglycemic agents are classified by their propen- peripheral glucose uptake and utilization.30 The
sity to cause side effects that have a negative glucose-lowering effect of metformin is highly
impact on DM disease state treatment5, 27 variable, with 35% of patients failing to achieve
(Table 4). initial decreases in hemoglobin HbA1c.31 About
1136

Table 3. Common MODY Subtypes, Phenotypes, and Select Associated Genetic Mutations
Implicated gene Frequency in
Subtypea (affected protein) Mutationsb Chromosome MODY patients Age at diagnosis Pathophysiology Phenotype
MODY 120–23 HNF-4a Naturally occurring, 20 5% Adolescence/Early Transcription Neonatal
(Hepatocyte Heterozygous (Q268X, adulthood factor (b-cell hyperinsulinism,
Nuclear Factor 4a) R154X, R127W) function in pancreas) diabetes
20–23
MODY 28, GCK (Glucokinase) Heterozygous (A378T, 7 10–60% Birth to early Hexokinase (glucose Mild hyperglycemia
E339K) childhood sensor in pancreas
and liver)
MODY 320–23 TCF1 or HNF-1a Heterozygous (291 + C) 12 30–60% Adolescence/Early Transcription factor Diabetes
(Hepatocyte adulthood (b-cell function in
Nuclear Factor 1a) pancreas and kidney)
MODY 420–23 IPF1 or PDX1 Heterozygous 13 <1% Early adulthood Transcription factor Diabetes
(Insulin Promoter (Pro63fsdelC) (b-cell function
Factor 1) in pancreas)
MODY 520 TCF1 or HNF-1b Heterozygous 17 3–10% Adolescence/Early Transcription factor Diabetes
(Hepatocyte (E101X, delT) adulthood (b-cell function
Nuclear Factor 1b) in pancreas)
MODY 620 NEUROD1 Heterozygous 2 Very rare Later in life Transcription factor Diabetes
(Neurogenic (206 + C) (b cell function in
Differentiation factor 1) pancreas and kidney)
23–26
TNDM20, ZAC, ABCC8, Heterozygous Rare Birth to 6 mo Transient
KCNJ11, HNF-1b diabetes
23–26
PNDM20, KCNJ11, ABCC8, Heterozygous, Rare Birth to 6 mo Permanent
GCK*, IPF1*, *Homozygous diabetes
MODY = maturity-onset diabetes of the young.
a
PHARMACOTHERAPY Volume 37, Number 9, 2017

MODY subtypes 1–14 have been characterized at the genetic level, but only subtypes 1–6 have clinically defined phenotypes. MODY 7–14 are very rare heterozygous single-point mutations
typically occurring in early childhood/adolescence.
b
Mutations are select representative variants listed for studies of affected patients in the Online Mendelian Inheritance in Man database at https://www.omim.org. Because MODY is a relatively
rare type of diabetes, not all mutations listed in the table are represented in all MODY patients.
PERSONALIZED MEDICINE IN DIABETES MANAGEMENT Elk and Iwuchukwu 1137
Table 4. Classification of Oral Medications Used in Diabetes Mellitus Treatment5, 27

Mechanism of Mechanism of HbA1c % Hypoglycemia Weight


Drug class Drugs action: cellular action: physiologic decrease risk effect
Biguanides* Metformin Activate AMPK, which Cause decrease in 1.0–1.5 Neutral Slight
then increase hepatic glucose loss
intracellular AMP production and intestinal
absorption of glucose;
improve insulin sensitivity
Sulfonylureas* Glyburide Close KATP channels Stimulate insulin release 0.8 Moderate Gain
Glipizide on pancreatic b-cell from b cells; reduce to severe
Glimepiride plasma membranes glucose output from
the liver, increase
insulin sensitivity
Meglitinides Repaglinide Close KATP channels Stimulate insulin release 0.7 Mild to Gain
Nateglinide on pancreatic b-cell from pancreatic b cells moderate
plasma membranes
Thiazolidinedione* Pioglitazone Activate the nuclear Improve target cell 0.8 Neutral Gain
Rosiglitazone transcription factor response to insulin
PPAR-c
a-Glucosidase Acarbose Inhibit intestinal Slow intestinal 0.6 Neutral Neutral
inhibitors Miglitol alpha-glucosidase carbohydrate
enzyme digestion/Absorption
DPP-4 inhibitors Sitagliptin Inhibit DPP-4 Glucose-dependent 0.7 Neutral Neutral
Saxagliptin activity, increasing increase in insulin
Linagliptin postprandial secretion and
Alogliptin active incretin decrease in glucose
(GLP-1, GIP) production
concentrations
SGLT2 inhibitors Canagliflozin Inhibit SGLT2 in the Block glucose 0.7–1.0 Neutral Loss
Empagliflozin proximal nephron reabsorption
in kidneys
GLP-1 Exenatide Activate GLP-1 Glucose-dependent 1 Neutral Loss
receptor agonists Liraglutide receptors increase in insulin
Albiglutide secretion and
Dulaglutide decrease in glucagon
secretion; slow gastric
emptying and increase
satiety
Amylin mimetics Pramlintide Activate amylin Decrease in glucagon 0.3 Severe Loss
receptors secretion; slow down
gastric emptying
and increase satiety
AMPK = AMP-activated protein kinase; DPP-4 = dipeptidyl peptidase-4; GIP = gastric inhibitory polypeptide; GLP-1 = glucagon-like peptide
1; KATP = ATP-sensitive K+ channel; PPAR-c = peroxisome proliferator-activated receptor gamma; SGLT2 = sodium-glucose cotransporter-2.
*Medications marked with an asterisk have evidence of a pharmacogenetic component.

a third of patients fail to achieve adequate glyce- taken up into the bloodstream by the organic
mic control on initial metformin monotherapy, cation transporter OCT1 (SLC22A1) that is also
and over time many patients become less sensi- responsible for its active hepatic uptake.28 Sub-
tive (responsive) to its glucose-lowering sequent secretion from the blood into the renal
effects.32 The genetic contribution to this vari- tubular cells is mediated by another organic
ability in response has been studied with a focus cation transporter, OCT2 (SLC22A2). Final
on the pharmacokinetics (PK) and pharmacody- excretion into the urine is mediated by two
namics (PD) of metformin. transporters in the multidrug and toxin extru-
sion family, MATE1 and MATE2 (SLC47A1 and
SLC47A2) (Figure 3).28
Metformin: Pharmacokinetics/Pharmacodynamics
Metformin is not metabolized upon ingestion;
Metformin: Evidence of Pharmacogenetic Associa-
rather, it is actively transported into various tis-
sues and cleared by renal excretion. Metformin tions
is taken up into the intestinal cells by plasma Although PD targets of metformin are not
monoamine transporters, or PMAT (SLC29A4), well established, the pharmacogenetics of the
1138 PHARMACOTHERAPY Volume 37, Number 9, 2017

Figure 3. Schematic presentation of pharmacogenomics of metformin. Metformin is absorbed in the small intestine via
plasma monoamine transporters (PMATs) encoded by SLC29A4 and is taken up into the bloodstream by an organic cation
transporter (OCT1) encoded by the SLC22A1 gene that is also responsible for active hepatic uptake. Metformin is secreted
from the blood to tubular cells by the organic cation transporter (OCT2) encoded by the SLC22A2 gene. Finally, metformin
is excreted into the urine via multidrug and toxin extrusion family transporters MATE1 and MATE2 that are encoded by
genes SLC47A1 and SLC47A2, respectively.

metformin PD-based response have been association between the minor C allele and met-
explored using both CGS and GWAS formin response,37 an association further con-
(Table 5).33 One of the most promising results firmed in meta-analyses of the three cohorts
was seen in the Genetics of DARTS (GoDARTS) alone and in combination with two previously
study. GoDARTs was a large-scale study with published studies.37 The Diabetes Prevention
1024 T2DM patients on metformin, a subset Program (DPP), another large T2DM cohort
from 3200 metformin-treated patients from the study, failed to show an association between this
parent U.K. Diabetes Audit and Research in top ATM variant and metformin effect in delay-
Tayside Scotland (DARTS) study.53 DARTS ing progression to diabetes from impaired glu-
aimed to identify all diabetes patients in the cose tolerance.40 These differences are most
Tayside community using electronic record likely due to the different phenotypes (predia-
linkage of multiple data sources and compare betes and overt diabetes) and study outcomes
ascertainment of cases with that of primary care (impact on diabetes incidence and ability to
registers.53 Subjects in the GoDARTs substudy reach treatment goals) in DPP and the other
consented to have their genomes interrogated.39 In studies.
this GWAS, the strongest association with PK studies seeking to explain differences in
metformin response was found with an intronic metformin exposure and response mainly focused
variant (rs11212617) near a candidate gene, on variations in genes coding for OCT1, OCT2,
ATM, or the ataxia telangiectasia mutated gene and MATE1.30, 33, 38, 54 Multiple genetic variants
that encodes a serine/threonine kinase.39 The in SLC22A1 have been associated with decreased
minor C allele of rs11212617 was associated metformin efficacy and increased renal clear-
with successful treatment outcomes on met- ance.34, 54 The GoDARTS study did not replicate
formin, defined as a reduction in HbA1c and an associations between two common SLC22A1 vari-
ability to reach treatment target HbA1c values ants (rs12208357, rs72552763) and glycemic
of 7% or lower.39 It has been postulated that response.35 The DPP study discovered a novel
mutations in the ATM gene may affect SLC22A1 variant (rs683369) associated with a
upstream regulation of AMPK and alter glyce- 31% decrease in diabetes incidence risk in met-
mic response to metformin.39 formin-treated patients and also validated
The rs11212617 when studied in three other SLC47A1 variants that were previously associated
different cohorts supported the positive with increased response to metformin (Table 5).33
Table 5. Select PK-PD Based Genotype-Phenotype Associations for Most Commonly Studied Oral Glucose Lowering Therapies in T2DM Patients
Drug Gene Variant Study outcomes Effect
Biguanides SLC22A1 (OCT1)33–36 rs622342 HbA1c 0.58% reduction in HbA1c for CC
(metformin) homozygotes
rs628031 (M408V) HbA1c No significant effect
rs35167514 (M420del) HbA1c No effect
rs12208357 (R61C) HbA1c No effect
metformin steady-state Gene dose effect: Decreasing AUC and
levels and decrease in pharmacodynamic effects at 6 and
HbA1c 24 mo corresponding
to number of OCT1 reduced function
alleles
rs12208357 (R61C), Gastrointestinal side 2-fold higher odds of gastrointestinal
rs35167514 (M420del) effects effects in individuals with OCT1
reduced function alleles
rs683369 T2DM prevention Nominal association with diabetes
prevention
rs622342 HbA1c No effect
37, 38
SLC22A2 (OCT2)33, rs316019 (G808T) Metformin steady-state No effect
levels and decrease in
HbA1c
rs662301 T2DM prevention Nominal association with diabetes
prevention
36, 37
SLC47A1 (MATE1)33, rs2289669 HbA1c Increased reduction in HbA1c levels
rs2252281 HbA1c Increased reduction inHbA1c levels for
homozygous CC patients
rs12943590 HbA1c Decreased in HbA1c reduction for
AA homozygotes
rs34399035 Metformin steady-state No effect
levels and decrease
in HbA1c
39, 40
ATM37, rs11212617 HbA1c Increased reduction in HbA1c and
increased treatment success
T2DM prevention No effect on time to progression to
diabetes

(continued)
PERSONALIZED MEDICINE IN DIABETES MANAGEMENT Elk and Iwuchukwu
1139
Table 5 (continued) 1140

Drug Gene Variant Study outcomes Effect


41–44
Sulfonylureas CYP2C9 rs1057910 Hypoglycemia Increased risk of hypoglycemia with
variant allele
rs1057910, rs1799853 Hypoglycemia Heterozygous and homozygous variants
more common in SU-treated patients
with hypoglycemia
rs1057910, rs1799853 Fasting blood glucose, Increased lowering of fasting plasma
SU dose requirements glucose, increased tolbutamide dose
reduction between dose 1 and 10 for
variant alleles
rs1057910, rs1799853 HbA1c Increased reduction in absolute HbA1c
levels
rs1057910, rs1799853 Hypoglycemia No overrepresentation of variant alleles
in SU-treated patients with
hypoglycemia
rs1057910, rs1799853 Time to stable dose No significant effects with variant alleles
37, 45–47
ABCC836, rs757110 HbA1c, fasting plasma G allele carriers had greater HbA1c
glucose reductions compared to the TT
homozygotes
HbA1c, fasting plasma Greater reductions in fasting plasma
glucose glucose and HbA1c with heterozygous
and homozygous variant alleles
Severe hypoglycemia No effect
KCNJ1141–44 rs5219 HbA1c, fasting plasma Significantly associated with greater
glucose decreases in fasting plasma glucose
HbA1c Greater HbA1c reduction in variant
allele carriers vs wild-type
homozygotes
Secondary failure
HbA1c, severe Variant allele associated with increased
hypoglycemia HbA1c levels and was less frequent in
SU-treated patients with severe
hypoglycemia vs controls
PHARMACOTHERAPY Volume 37, Number 9, 2017

SU response No effect
49
TCF7L248, rs12255372, rs7903146 HbA1c <7% Increased risk of treatment failure with
variants alleles
rs7903146 HbA1c <7% Increased risk of treatment failure with
variant allele
HbA1c and fasting Less HbA1c reduction and fasting plasma
plasma glucose glucose levels with variant alleles
reduction compared with wild-type alleles

(continued)
PERSONALIZED MEDICINE IN DIABETES MANAGEMENT Elk and Iwuchukwu 1141

Sulfonylureas

both SNPs, Less reduction in HbA1c for


decreases in fasting plasma glucose and

Greater HbA1c reduction but not weight


gain after rosiglitazone. No association

gain after rosiglitazone. No association


plasma levels of rosiglitazone, reduced
therapeutic response, and a lower risk

Decrease in fasting plasma glucose for


Less reduction in HbA1c, less weight
Sulfonylureas (SUs) are insulin secretagogues

Variant alleles associated with lower

homozygote variants of rs2241766


shown to be efficacious in the treatment of
Variant allele carriers had greater

T2DM by producing favorable lowering effect of


HbA1c and are recommended to be used in com-
bination with other agents for better blood glu-
Effect

of developing edema cose control.55 SUs bind to their target site on

AUC = area under the curve; Hb = hemoglobin; rs = reference sequence; SNP = single nucleotide polymorphism; SU = sulfonylurea; T2DM = type 2 diabetes mellitus.
pancreatic b islet cells, the sulfonylurea receptor
with pioglitazone

with pioglitazone
type 1 (SUR1), encoded by ABCC8, a member of
the ATP binding cassette gene superfamily.56, 57
When SUs bind to SUR1, it results in closing of
HbA1c

the pancreatic b-cell KATP channel, causing


membrane depolarization and triggering calcium
influx with subsequent exocytosis of insulin-
containing vesicles and increased insulin
secretion.57
Fasting plasma glucose,

HbA1c, fasting plasma


Rosiglitazone steady-
state levels, HbA1c,
Study outcomes

HbA1c, weight gain

HbA1c, weight gain


HbA1c reduction

Sulfonylureas: Pharmacokinetics/Pharmacodynamics
edema risk

The PK and PD responses of SUs are well


characterized. SUs undergo extensive and vari-
glucose

able hepatic metabolism by cytochrome P450


2C9 (CYP2C9). Polymorphisms in CYP2C9 were
shown to be one of the reasons behind the vari-
ability in elimination rates for most SUs.58 Two
CYP2C9 reduced function variants of interest,
rs11572080, rs10509681

CYP2C9*2 (rs1799853) and CYP29*3


rs2241766, rs1501299

(rs1057910), are associated with decreased clear-


ance and increased area under the curve (AUC)
Variant

of SUs in healthy volunteers.36


Response to SU therapy depends on b-cell
rs11572080
rs1801282

rs4149056

function, PK differences among patients, and


current T2DM status. Interpatient variability in
response to SU treatment is observed with fail-
ure of therapy and the incidence of hypo-
glycemia. Primary failure with SUs is reported in
10–20% of patients and secondary failure in 5–
7% of patients.56 Evidence from trials shows that
hypoglycemia develops in 31% of patients within
the first year of starting SU, progressing to 38%
of patients at 5 years of therapy.31, 59 Accord-
Gene

ing to one meta-analysis, the incidence of severe


SLCO1B151

hypoglycemia in SU-treated patients was 0.8%.60


ADIPOQ52
CYP2C851
50
PPAR-c

Sulfonylureas: Evidence of Pharmacogenetic Associ-


ations
The largest pharmacogenetics-based clinical
Table 5 (continued)

Thiazolidinediones

study to date was the GoDARTS study with


1073 patients, half of whom were on SU
monotherapy and half on dual therapy (SU and
metformin).41 In this study, patients with two
copies of the CYP2C9*2 or *3 alleles were 3.4
Drug

times more likely to achieve target treatment


1142 PHARMACOTHERAPY Volume 37, Number 9, 2017

HbA1c levels of less than 7% compared with (rs757110) in ABCC8, two tightly linked vari-
patients with two copies of the wild-type ants occurring together as a haplotype due to
CYP2C9*1 allele. Variant allele carriers were less linkage.62 This haplotype was shown to be less
likely to experience treatment failure with SU sensitive to SU treatment in vitro.62 Clinical
monotherapy.41 Reports were published on asso- studies probing associations of these variants
ciations between CYP2C9 genotypes and the risk with either therapeutic response or occurrence
of hypoglycemia.42, 43 An initial study showed of hypoglycemia have proven equivocal with
an odds ratio of 5.2 for the *2/*3 and the *3/*3 some studies showing a positive association and
genotypes for risk of SU-induced severe hypo- others showing no association.45, 47, 61, 63, 64 Of
glycemia,43 but a subsequent 10-year follow-up note is the fact that studies differed with respect
study did not corroborate these findings.44 to SUs used, dose, study outcomes, ethnicities,
Although confirming the role of CYP2C9 in SU cohort characteristics, and sample size.
metabolism, the GoDARTS study and other stud- With the advent of whole-genome sequencing
ies41, 43, 44 also emphasize that determination of technologies, novel genetic loci associated with
CYP2C9 genotype may not be sufficient itself to T2DM and SU response have been discovered;
predict SU response. Translating these PK gene- the most replicated is the TCF7L2 loci.16, 48, 49
based studies to clinical outcomes, such as fast- In 901 SU-treated T2DM patients identified from
ing serum glucose levels and hypoglycemia, GoDARTS, an intronic SNP, rs12255732, in
showed only modest effects due most likely to TCF7L2 was strongly associated with SU
limited sample size and lack of statistical power. response.48 TCF7L2 is expressed in pancreatic b
For the PD of SUs, initial CGS studies focused cells, and reduced function alleles have been
on the ABCC8 and KCNJ11 genes encoding associated with lower insulin secretion sugges-
SUR1 and Kir6.2, the different subunits of the tive of a direct or indirect role on b-cell func-
pancreatic b-cell KATP channels based on a tion.48 Individuals with two copies of the variant
priori knowledge of their association with dia- T allele were two times less likely to reach target
betes (Figure 4).45, 46, 61 Results were consistent HbA1c levels below 7% within 1 year of treat-
for variants previously associated with T2DM: ment compared with those with two copies of
E23K (rs5219) in KCNJ11 and S1369A the G variant of rs12255732.48 These findings

Figure 4. Schematic diagram of pharmacogenomics of sulfonylureas. Sulfonylureas bind to sulfonylurea receptor type 1
(SUR1) located on the pancreatic b islet cell that is encoded by the ABCC8, a member of the ATP-binding cassette gene
superfamily. When SUs bind to SUR1, it results in closing of the pancreatic b-cell potassium channel (KATP), made up of
Kir6.2 subunits encoded by KCNJ11. KATP channel closing causes membrane depolarization and triggering calcium influx
with subsequent exocytosis of insulin-containing vesicles and increased insulin secretion, a process postulated to be regulated
by TCF7L2. Insulin binds to insulin receptor site (IRS-1) encoded by IRS-1 and located on the surface of insulin-sensitive
tissue. IRS-1 gets phosphorylated and regulates the function of insulin inside the cell.
PERSONALIZED MEDICINE IN DIABETES MANAGEMENT Elk and Iwuchukwu 1143

were confirmed by other independent stud- it was hypothesized that the lower metabolic
ies.49, 65 Another intron variant in TCF7L2, activity CYP2C8*2 and *3 variants would result
rs7903146, was associated with lower response in decreased metabolism and increased drug
and increased risk of treatment failure for carri- levels, clinical studies did not corroborate these
ers of the T allele compared with the CC geno- assumptions. Instead, they showed higher clear-
types.48, 49 Other PD targets such as Insulin ance and lower AUC for subjects with two
receptor substrate-1 encoded by the IRS-1 gene copies of the *3 alleles compared with the wild
have been associated with T2DM and SU type.72 The CYP2C8*3 allele is relatively rare in
response. The most common variant in IRS-1, whites, and adjustments for baseline variables
rs1801278, was associated with an increased risk may have reduced the impact of genetic variabil-
of secondary failure to SU therapy in two sepa- ity on rosiglitazone PK. Furthermore, TZDs are
rate cohorts of T2DM patients.66, 67 substrates of OATP1B (SLCO1B1), the main
organic anion transporter protein responsible for
Thiazolidinediones their active shuttling into hepatocytes.71 A joint
investigation on the effects of CYP2C8 and
Although their use is limited due to risk of SLCO1B1 polymorphisms was recently con-
severe adverse events such as heart failure, ducted in 833 Scottish patients with T2DM trea-
myocardial infarction, and bladder cancer, thia- ted with TZDs.51 Results showed that although
zolidinediones (TZDs) still have a role in dia- the CYP2C8*3 variant was associated with
betes pharmacotherapy. Decision to use a TZD reduced glycemic response to rosiglitazone, the
in a patient is made after careful risk versus ben- common SCLO1B1 521T>C variant, in contrast,
efit evaluation. TZDs are insulin sensitizers that was associated with an enhanced glycemic
increase insulin-dependent glucose elimination, response.51 A subset of patients known as super-
reduce hepatic glucose production, and decrease responders based on combined CYP2C8 (*1/*1)
circulating free fatty acids. TZDs act by activat- and SCLO1B1 (CC) genotypes had a 0.39%
ing PPAR-c; the activated receptor then binds to greater HbA1c reduction with rosiglitazone com-
DNA response elements and controls the expres- pared with poor responders.51
sion of genes involved in maintenance of meta- Studies on TZDs and PD targets have shown
bolic homeostasis.68 PPAR-c activation in some progress. Many studies to date have
mature adipose cells induces expression of genes focused on polymorphisms in the PPAR-c gene,
involved in the insulin signaling cascade and as a logical first choice because it encodes the
a result improves insulin sensitivity in diabetic binding site for TZD ligands (Figure 5). The
patients. In addition, these genes were demon- PPAR-c gene contains a common nonsynony-
strated to reduce insulin resistance.69 mous SNP, rs1801282, that gives rise to a codon
As seen with the other OGLTs, there is pro- 12 proline to alanine substitution (Pro12Ala)
nounced interindividual variability in response implicated in more than 50 studies on diabetes
to TZD treatment. Clinical studies of pioglita- susceptibility.73 Meta-analyses of these associa-
zone and rosiglitazone show that between 12% tions show an almost 20% reduction in T2DM
and 45% of T2DM patients failed to achieve suf- risk for Ala12 allele carriers compared with car-
ficient reduction in either fasting plasma glucose riers of the Pro12 allele.73, 74 Clinical transla-
and/or HbA1c concentrations.70 tional studies probed for associations with the
Ala12 allele and response to TZD therapy in at-
Thiazolidinediones: Pharmacokinetics/Pharmacody- risk patients and patients with T2DM. Reports
namics and Evidence of Pharmacogenetic Associa- on the Ala12 allele and TZD response in dia-
betes patients are conflicting. Whereas one study
tions
showed no difference in frequency of subjects
TZDs and the impact of variation in PK genes with one copy of the Ala12 allele,75 another
on response has been evaluated. TZDs are lar- study reported a higher frequency of the Ala12
gely metabolized by the polymorphic cyto- allele in responders to pioglitazone.76 One study
chrome P450 2C8 (CYP2C8) and to a much reported a significantly higher response to
lesser extent by CYP2C9 and CYP3A4.71 Genetic rosiglitazone at 12 weeks in Ala12 subjects com-
variation in CYP2C8 was reported to influence pared with homozygous Pro12 subjects (43.72%
the PK of rosiglitazone with significantly lower vs 86.67%).50 Again, key differences among
AUC and higher oral clearance for *1/*3 sub- studies that may have contributed to inconsis-
jects compared with *1/*1 subjects.71 Although tent results include differences in TZD type,
1144 PHARMACOTHERAPY Volume 37, Number 9, 2017

Figure 5. Schematic presentation of pharmacogenomics of thiazolidinediones. (A) Thiazolidinediones (TZDs) are substrates
of OATP1B, the organic anion transporter protein encoded by SLCO1B1. OATP1B1 is the main hepatic transporter
responsible for the active shuttling of organic anion compounds like TZDs into hepatocytes. (B) TZDs act by activating the
PPAR-c receptor encoded by the PPAR-c gene; the activated receptor then binds to DNA response elements and controls the
expression of genes involved in maintenance of metabolic homeostasis. (C) Adiponectin encoded by the ADIPOQ gene is a
protein hormone expressed abundantly in adipose tissue where it promotes glucose uptake and fatty acid oxidation.
Adiponectin actions are controlled by PPAR-c receptor activation.

treatment duration, inclusion criteria, baseline studies have yet to be replicated. Other PD
metabolic conditions, and ethnicity. genes linked to TZDs have been studied without
Another non-PPAR-c TZD target gene of inter- any significant replication reports.
est is the ADIPOQ gene that encodes adiponectin
(Figure 5). Low levels of adiponectin have been
Gliptins
associated with insulin resistance, and TZDs are
known to increase adiponectin levels that may Gliptins, also known as dipeptidyl peptidase
contribute in part to their insulin-sensitizing IV (DPP4) inhibitors, are classified as incretin
effects.74 Common exonic (45T>G; rs16861194) mimetics. They are newer agents that exert their
and intronic (276G>T; rs1501299) SNPs in ADI- effects by inhibiting a key enzyme in the incretin
POQ have been studied. Carriers of the 45G signaling pathway, preventing the degradation of
variant and the 276G wild-type alleles, whether endogenous glucagon-like peptide-1 (GLP-1)
studied alone or in haplotype combinations, had receptor agonist and gastric inhibitory peptide
smaller decreases in fasting plasma glucose and (GIP).79 GLP-1 is secreted by the intestines after
HbA1c compared with patients carrying the intake of food and acts to induce insulin secre-
other alleles.52 In addition to studies probing tion from pancreatic b cells, suppresses the
drug response, occurrence of the many troubling release of glucagon, inhibits gastric emptying,
and limiting adverse effects with TZDs also led and reduces appetite. Inhibiting the degradation
to genetic explorations of any associated mecha- of GLP-1 leads to prolonged secretion of insulin
nisms. One study reported an association and inhibition of glucagon.79
between weight gain and a SNP in the perilipin
gene.77 A genetic substudy of the Diabetes
Gliptins: Evidence of Pharmacogenetic Associations
Reduction Assessment with Ramipril and
Rosiglitazone Medication (DREAM) study In terms of their metabolic profile, DPP-4
showed a significant association with rosiglita- inhibitors are not typical substrates for either
zone-induced edema and rs6123045 in the cytochrome P450 (except saxagliptin, metabo-
nuclear factor of the activated T-cells cytoplas- lized by CYP3A4/A5) or any major transporter
mic calcineurin-dependent 2 (NFATC2) gene in families because their main mode of clearance is
subjects of European descent.78 Neither of these by renal excretion.7 Due to the relatively recent
PERSONALIZED MEDICINE IN DIABETES MANAGEMENT Elk and Iwuchukwu 1145

entry of these drugs into the market, very few when selecting a diabetic agent and using a per-
pharmacogenetic studies have been conducted.80 sonalized approach in DM management. Despite
Studies on genetic influence on insulin secretion the multiple factors that clinicians need to con-
after administration of human GLP-1 to healthy sider when selecting a treatment strategy for DM
individuals may be extrapolated to explain today, there is still room for incorporating phar-
observed differences in response to GLP-1 recep- macogenomics for a more personalized manage-
tor agonists and DPP-4 inhibitors. A recent ment of DM (Figure 1).
study showed a SNP, rs7202877, near the
CTRB1 and CTRB2 loci influenced the response
Using Genetics for the Prediction of DM
to DPP-4 inhibitor treatment in patients with
T2DM.81 The CRTB locus encodes for the diges- Primary studies and meta-analyses of genetics
tive enzyme chymotrypsin, an important regula- studies on diabetes risk have yielded many
tor of the incretin pathway.79 The G allele of genetic associations that increase our under-
rs7202877 was previously associated with an standing of T2DM pathophysiology. Although
increased risk for T1DM and a decreased risk these explorations only explain a small fraction
for T2DM.14 In one study, the rs7202877G allele of genetic contribution to phenotype, the associ-
was associated with enhanced GLP-1 secretion ations can be used to improve disease classifica-
in healthy individuals and reduced response to tion. One example of this is with the use of
DPP-4 inhibitor treatment in patients with genetic risk scores (GRS) to predict the risk of
T2DM. diabetes incidence. GRS scores are typically
weighted by participants, and the number of risk
alleles per SNP are summed over the total num-
Personalized Treatment Strategy for DM
ber of representative SNPs associated with the
Managing patients with DM presents a chal- risk of diabetes. The Framingham Heart Study
lenge. Treatment failure, risk of hypoglycemia, (FHS) and the Diabetes Prevention Program
and selection of appropriate agents based on (DPP) are representative cohorts that have suc-
pathogenesis of the disease creates a need for cessfully used this approach to predict diabetes
using precision medicine in DM. With so many risk.83, 84 In the FHS, a GRS based on 18 risk
different classes of medications available, the alleles predicted new cases of diabetes with a
guidelines recommend use of patient-specific slightly better prediction of risk than common
approaches when designing DM treatment risk factors (such as body mass index, hyperten-
plans.55 Clinicians should select a treatment sion, family history of diabetes, ethnicity, and
strategy and goals of therapy that provide the age) alone.84 In the DPP, a high GRS (based on
most benefit with the least harm. To address the 34 risk alleles) was associated with an increased
question of efficacy among commonly used dia- risk of developing diabetes but was subsequently
betes medications in various patient populations, attenuated by lifestyle interventions in the ana-
a national randomized interventional study was lytical model.83 Although these examples pro-
proposed.82 The Glycemia Reduction Approaches vide an illustration of how pharmacogenomics
in Diabetes: A Comparative Effectiveness Study can be incorporated into the risk prevention
(GRADE) is currently enrolling adult patients paradigm, the challenge of selecting the most
with a history of DM for less than 10 years and relevant and clinically validated risk variants is
currently on metformin monotherapy. Patients one that is yet to be overcome.
are divided into four groups to receive met-
formin with either glimepiride, sitagliptin, Applications of Pharmacogenomics in
liraglutide, or basal insulin glargine. The primary
Monogenic Diabetes Syndrome
outcome measure is time to primary failure of
treatment defined by a HbA1c higher than 7% For disease management, the most extensive
over a period of 4–7 years. Investigators will application of pharmacogenetics in DM to date
compare personalized response to treatment, has been with the monogenic forms of the dis-
looking at adverse events, tolerability, microvas- ease. Identification of the correct MODY subtype
cular and macrovascular complications, quality has a tremendous impact on treatment. It is rec-
of life, and cost-effectiveness. Participants’ phe- ommended that patients presenting with diabetes
notypes will be studied to help guide selection of in the first 6 months of life be tested for muta-
optimal diabetic agent(s).82 The results of tions in KCNJ11 and ABCC8 to rule out
GRADE should provide additional guidance TNDM.23 The identification of the NDM subtype
1146 PHARMACOTHERAPY Volume 37, Number 9, 2017

is significant due to associated mutations occur- Monogenic Diabetes Registry of MODY patients
ring in the pancreatic b-cell KATP channels and is useful for further guidance on current
their role in guiding treatment choice. SU action research and treatment strategies.87
on these channels causes increased insulin secre-
tion, making them the most ideal agents for Pharmacogenomics in Predicting Response to
NDM management. TNDM patients compared
Therapy
with those with PNDM are likely to have signifi-
cantly higher endogenous insulin production, With respect to the pharmacogenetics of
making them much more responsive to SUs.23 OGLTs, metformin is still the guideline-based
They can thus be successfully treated with low- first-line monotherapy for diabetes. The variabil-
dose SUs in place of standard insulin therapy ity in response and treatment failure with met-
versus higher SU doses for PNDM. The switch formin is clearly indicative of a differential effect
from insulin to SU should only be made after a that may be genetically induced. Variants in
molecular genetic diagnosis. There are estab- metformin PK and transporter genes have been
lished translational research diagnosis and treat- the most widely studied and have greatly
ment approach guidance/protocols for MODY advanced our knowledge of metformin pharma-
and NDM as well as a MODY probability calcu- cogenomics. Replicated SLC transporter variants
lator for clinicians’ reference.85 could play a role in guideline development for
Patients with MODY 1 and 3 are prime targets metformin use in select patient populations
for personalized therapy as embodied by the based on combined GRS. Pharmacogenomics-
reported relatively safe transition from insulin to guided therapy based on a greater understanding
SU monotherapy in these patients. The finding of metformin genes and pathways could help
that these patients are generally hypersensitive clinicians if outcomes (patient response and
to SUs makes such a transition possible. This probable treatment failures) could be determined
hypersensitivity to SUs has been attributed to a priori. Alternative guideline-based recommen-
downregulation of HNF1-a and HNF4-a target dations can then be used. This type of pharma-
genes in the liver, leading to decreased hepatic cogenomics-guided approach could lead to
uptake of SUs and increased levels in the general long-term dollar cost savings compared with a
circulation.80 “wait until treatment fails then switch”
Personalized treatment strategy selection may approach.
not always include pharmacotherapy, a paradigm Response to SU therapy depends on b-cell
seen with the appropriate identification of function and its insulin production ability.
patients with MODY 2, a frequently misdiag- Results with clinical studies of the CYP2C9 gene
nosed subset.8 These patients present with mild and SU response have been promising as seen in
hyperglycemia, which improves on its own, and the GoDARTs study. Variations in KCNJ11 and
generally do not progress to overt diabetes with ABCC8 genes were also identified as associated
associated macrovascular and microvascular with a high response to SU treatment in certain
complications. Consequently, patients with patients with hyperglycemia.46 These critical
MODY 2 typically have no need for pharma- findings may potentially lead to testing/screening
cotherapy and can be managed by diet alone.8 patients for these polymorphisms. Advance
Ideally, clinicians ought to be aware that knowledge of these and other validated strong
patients with an onset of diabetes before age of genetic signals, such as those in TCF7L2, may
25 should be referred for genetic testing to rule also assist in careful dose titration for SU-sensi-
out MODY and prevent misdiagnosis. The Inter- tive versus SU-tolerant patients. On a pharmaco-
national Society for Pediatric and Adolescent genetic basis, SU therapy can therefore be
Diabetes (ISPAD) guidelines provide very initiated as the most effective choice in “high-
detailed recommendations about when to sus- response” patients and avoided in “low/no-
pect the presence of MODY.23 The American response” patients similar to the current success
Diabetes Association guidelines also provide rec- with some MODY patients.
ommendations for the screening of MODY in With the TZDs, although results from many
children and adolescents initially diagnosed with of the pharmacogenomic studies conducted
DM within the first 6 months of life.5 Genetic were clinically significant, the current guideli-
tests for MODY-related genes and CLIA- nes stipulate consideration of risk versus benefit
approved labs for such testing are available on before initiating use. It is doubtful there will
the Genetic Testing Registry.86 The National be any additional benefit to incorporation of
PERSONALIZED MEDICINE IN DIABETES MANAGEMENT Elk and Iwuchukwu 1147

pharmacogenomics based on their current role future, knowing which patients are likely to
in treatment. develop severe side effects can help with strati-
It is our opinion that although there are cer- fied medicine.
tainly advances in genetic involvement and asso-
ciations with OGLTs, there are as yet no strong
Conclusion
clinical guidelines or recommendations for
genetic testing and application in clinical With so many different classes of medications
practice. available for managing diabetes, the recommenda-
tion from the guidelines is to use a patient-speci-
Future Prospects of Using Genetics for Therapy fic approach and to select treatment strategies
providing the most benefit with the least harm.
of DM
Using personalized medicine in the treatment of
Common risk factors for T2DM include obe- diabetes where patients’ diabetes types can be
sity, dyslipidemia, family history, age, low physi- precisely identified with respect to pathology fits
cal activity, Hispanic and African-American perfectly with the selection of agents that will best
ethnicity, smoking, and unhealthy diet.5 Compos- treat specific pathologies of such stratified dia-
ite GRS may better predict diabetes risk in combi- betes. To make it even more patient specific,
nation with other risk factors but would not be pharmacogenomics may be used to identify
expected to be an efficient discriminator due to variations in medication disposition. Based on the
low effect sizes of single common variants in evidence, attaining glycemic goal(s) is not enough
models. However, GRS may be useful in younger to prevent diabetes-related complications. Con-
patients who have not developed obvious risk trolling comorbidities and most importantly pre-
factors and may also prove useful for newborn vention of diabetes onset and progression needs
diabetes susceptibility genetic screening and fol- to be addressed.
low-up.83, 84 Another utility for GRS would be in Overall, data from pharmacogenomics-based
ethnically focused screening and targeted treat- studies may help present a clearer picture of any
ment based on the presence or absence of known implicated molecular mechanisms en route to
susceptibility genes because reports of popula- selecting pharmacoeconomically viable and rele-
tion-specific T2DM susceptibility variants con- vant treatment strategies for the complex multi-
tinue to emerge from large consortia studies.17 factorial disease that is DM.
Genetics-based association studies in OGLTs
and their role in DM treatment identified herita- References
ble components of a patient’s response to therapy. 1. World Health Organization. Diabetes: Key Facts, November
Although at this time not enough evidence for 2016. Available from www.who.int/mediacentre/factsheets/
clinical utility exists, future larger cohorts and fs312/en/. Accessed February 13, 2017.
2. World Health Organization. Diabetes: Global report on dia-
combinations from academic, practice, and indus- betes. Available from www.who.int/diabetes/global-report/en/.
try DM research groups may aid in identifying Accessed February 13, 2017.
more genes implicated in OGLT response. The 3. Centers for Disease Control and Prevention. Chronic Disease
Prevention and Health Promotion: Diabetes, July 2016. Avail-
large evidence base generated from these high- able from www.cdc.gov/chronicdisease/resources/publications/
powered consortia studies may be an impetus for aag/diabetes.htm. Accessed February 13 2017.
development of guidelines for the use of genetic 4. American Diabetes Association. Economic costs of diabetes in
the U.S. in 2012. Diabetes Care 2013;36:1033–46.
information in diabetes pharmacotherapy. 5. American Diabetes Association. Standards of medical care in
Use of pharmacogenetics for the prediction diabetes. Diabetes Care 2016;39:S1–106.
and prevention of side effects from OGLTs may 6. Barroso I. Genetics of type 2 diabetes. Diabet Med 2005;22:
517–35.
be an idea whose time is around the corner. 7. Hupfeld CJ, Courtney H, Olefsky JM. Type 2 diabetes melli-
Metformin and SUs, although mainline agents tus: etiology, pathogenesis, and natural history. In: Jameson JL,
for therapy, are more prone to cause intolerant De Groot LJ, eds. Endocrinology: adult and pediatric. Philadel-
phia, PA: Elsevier Health Sciences, 2010:765–87.
gastrointestinal effects or hypoglycemia com- 8. Carmody D, Naylor RN, Bell CD, et al. GCK-MODY in the
pared with other agents. As such, testing US National Monogenic Diabetes Registry: frequently misdiag-
patients a priori for any implicated clinically val- nosed and unnecessarily treated. Acta Diabetol 2016;53:703–8.
9. Tan JT, Chia KS, Ku CS. The molecular genetics of type 2 dia-
idated metformin or SU intolerance or low- betes: past, present and future. In: Encyclopedia of life
response susceptibility variants could be an sciences. Chichester, UK: John Wiley & Sons, 2009. doi: 10.
adjunct tool to maximizing their use in therapy. 1002/9780470015902.a0021994.
10. Denny JC, Ritchie MD, Basford MA, et al. PheWAS: demon-
The same extrapolations could also be made for strating the feasibility of a phenome-wide scan to discover
restricted OGLTs such as rosiglitazone. In the gene-disease associations. Bioinformatics 2010;26:1205–10.
1148 PHARMACOTHERAPY Volume 37, Number 9, 2017
11. Brunetti A, Chiefari E, FTI D. Recent advances in the molecu- response to metformin and lifestyle intervention in the dia-
lar genetics of type 2 diabetes mellitus. World J Diabetes betes prevention program. Diabetes 2010;59:2672–81.
2014;5:128–40. 34. Shu Y, Sheardown SA, Brown C, et al. Effect of genetic varia-
12. Owen KR, McCarthy MI. Genetics of type 2 diabetes. Curr tion in the organic cation transporter 1 (OCT1) on metformin
Opin Genet Dev 2007;17:239–44. action. J Clin Invest 2007;117:1422–31.
13. Doria A, Patti M-E, Kahn CR. The emerging genetic architec- 35. Zhou K, Donnelly LA, Kimber CH, et al. Reduced-function
ture of type 2 diabetes. Cell Metab 2008;8:186–200. SLC22A1 polymorphisms encoding organic cation transporter
14. Morris AP, Voight BF, Teslovich TM, et al. Large-scale associ- 1 and glycemic response to metformin: a GoDARTS study.
ation analysis provides insights into the genetic architecture Diabetes 2009;58:1434–9.
and pathophysiology of type 2 diabetes. Nat Genet 2012;44: 36. Becker ML, Pearson ER, Tkac I. Pharmacogenetics of oral
981–90. antidiabetic drugs. Int J Endocrinol 2013;2013: Article ID
15. The Wellcome Trust Case Control Consortium. Genome-wide 686315. doi: 10.1155/2013/686315.
association study of 14,000 cases of seven common diseases 37. van Leeuwen N, Nijpels G, Becker ML, et al. A gene variant
and 3,000 shared controls. Nature 2007;447:661–78. near ATM is significantly associated with metformin treatment
16. Grant SF, Thorleifsson G, Reynisdottir I, et al. Variant of response in type 2 diabetes: a replication and meta-analysis of
transcription factor 7-like 2 (TCF7L2) gene confers risk of five cohorts. Diabetologia 2012;55:1971–7.
type 2 diabetes. Nat Genet 2006;38:320–3. 38. Chen Y, Li S, Brown C, et al. Effect of genetic variation in the
17. Fuchsberger C, Flannick J, Teslovich TM, et al. The genetic organic cation transporter 2 on the renal elimination of met-
architecture of type 2 diabetes. Nature 2016;536:41–7. formin. Pharmacogenet Genomics 2009;19:497–504.
18. GWAS Catalog. The NHGRI-EBI catalog of published gen- 39. Zhou K, Bellenguez C, Spencer CC, et al. Common variants
ome-wide association studies, February 2017. Available from near ATM are associated with glycemic response to metformin
www.ebi.ac.uk/gwas/. Accessed February 13, 2017. in type 2 diabetes. Nat Genet 2011;43:117–20.
19. Bonnefond A, Froguel P. Rare and common genetic events in 40. Florez JC, Barrett-Connor E, Jablonski KA, et al. The C allele
type 2 diabetes: what should biologists know? Cell Metab of ATM rs11212617 does not associate with metformin
2015;21:357–68. response in the diabetes prevention program. Diabetes Care
20. Vaxillaire M, Froguel P. Genetic basis of maturity-onset dia- 2012;35:1864–7.
betes of the young. Endocrinol Metab Clin North Am 2006;35: 41. Zhou K, Donnelly L, Burch L, et al. Loss-of-function CYP2C9
371–84. variants improve therapeutic response to sulfonylureas in type
21. Chambers C, Fouts A, Dong F, et al. Characteristics of matu- 2 diabetes: a Go-DARTS study. Clin Pharmacol Ther 2010;87:
rity onset diabetes of the young in a large diabetes center. 52–6.
Pediatr Diabetes 2016;17:360–7. 42. Ragia G, Petridis I, Tavridou A, Christakidis D, Manolopou-
22. Pihoker C, Gilliam LK, Ellard S, et al. Prevalence, characteris- los VG. Presence of CYP2C9*3 allele increases risk for hypo-
tics and clinical diagnosis of maturity onset diabetes of the glycemia in type 2 diabetic patients treated with sulfonylureas.
young due to mutations in HNF1A, HNF4A, and glucokinase: Pharmacogenomics 2009;10:1781–7.
results from the SEARCH for Diabetes in Youth. J Clin Endo- 43. Holstein A, Plaschke A, Ptak M, et al. Association between
crinol Metab 2013;98:4055–62. CYP2C9 slow metabolizer genotypes and severe hypogly-
23. Rubio-Cabezas O, Hattersley AT, Njølstad PR, et al. ISPAD caemia on medication with sulphonylurea hypoglycaemic
Clinical Practice Consensus Guidelines 2014. The diagnosis agents. Br J Clin Pharmacol 2005;60:103–6.
and management of monogenic diabetes in children and ado- 44. Holstein A, Hahn M, Patzer O, Seeringer A, Kovacs P, Stingl
lescents. Pediatr Diabetes 2014;15(Suppl 20):47–64. J. Impact of clinical factors and CYP2C9 variants for the risk
24. Babenko AP, Polak M, Cave H, et al. Activating mutations in of severe sulfonylurea-induced hypoglycemia. Eur J Clin Phar-
the ABCC8 gene in neonatal diabetes mellitus. N Engl J Med macol 2011;67:471–6.
2006;355:456–66. 45. Feng Y, Mao G, Ren X, et al. Ser 1369Ala variant in sulfony-
25. Gloyn AL, Pearson ER, Antcliff JF, et al. Activating mutations lurea receptor gene ABCC8 Is associated with antidiabetic effi-
in the gene encoding the ATP-sensitive potassium-channel cacy of gliclazide in Chinese type 2 diabetic patients. Diabetes
subunit Kir6.2 and permanent neonatal diabetes. N Engl J Care 2008;31:1939–44.
Med 2004;350:1838–49. 46. Florez JC, Jablonski KA, Kahn SE, et al. Type 2 diabetes-asso-
26. Colombo C, Delvecchio M, Zecchins C, et al. Transient ciated missense polymorphisms KCNJ11 E23K and ABCC8
neonatal diabetes mellitus is associated with a recurrent A1369S influence progression to diabetes and response to
(R201H) KCNJ11 (KIR6.2) mutation. Diabetologia 2005;48: interventions in the Diabetes Prevention Program. Diabetes
2439–41. 2007;56:531–6.
27. Canadian Diabetes Association Clinical Practice Guidelines 47. Holstein JD, Kovacs P, Patzer O, Stumvoll M, Holstein A.
Expert Committee. Pharmacologic management of type 2 dia- The Ser1369Ala variant of ABCC8 and the risk for severe sul-
betes: 2016 interim update. Can J Diabetes 2016;40:193–5. fonylurea-induced hypoglycemia in German patients with type
28. Gong L, Goswami S, Giacomini KM, Altman RB, Klein TE. 2 diabetes. Pharmacogenomics 2012;13:5–10.
Metformin pathways: pharmacokinetics and pharmacodynam- 48. Pearson ER, Donnelly LA, Kimber C, et al. Variation in
ics. Pharmacogenet Genomics 2012;22:820–7. TCF7L2 influences therapeutic response to sulfonylureas: a
29. Zhou G, Myers R, Li Y, et al. Role of AMP-activated protein GoDARTs study. Diabetes 2007;56:2178–82.
kinase in mechanism of metformin action. J Clin Invest 49. Holstein A, Hahn M, Korner A, Stumvoll M, Kovacs P.
2001;108:1167–74. TCF7L2 and therapeutic response to sulfonylureas in patients
30. Pawlyk AC, Giacomini KM, McKeon C, Shuldiner AR, Florez with type 2 diabetes. BMC Med Genet 2011;12:30.
JC. Metformin pharmacogenomics: current status and future 50. Kang ES, Park SY, Kim HJ, et al. Effects of Pro12Ala polymor-
directions. Diabetes 2014;63:2590–9. phism of peroxisome proliferator-activated receptor gamma2
31. Kahn SE, Haffner SM, Heise MA, et al. Glycemic durability of gene on rosiglitazone response in type 2 diabetes. Clin Phar-
rosiglitazone, metformin, or glyburide monotherapy. N Engl J macol Ther 2005;78:202–8.
Med 2006;355:2427–43. 51. Dawed AY, Donnelly L, Tavendale R, et al. CYP2C8 and
32. Riedel AA, Heien H, Wogen J, Plauschinat CA. Loss of glyce- SLCO1B1 variants and therapeutic response to thiazolidine-
mic control in patients with type 2 diabetes mellitus who were diones in patients with type 2 diabetes. Diabetes Care
receiving initial metformin, sulfonylurea, or thiazolidinedione 2016;39:1902–8.
monotherapy. Pharmacotherapy 2007;27:1102–10. 52. Kang ES, Park SY, Kim HJ, et al. The influence of adiponectin
33. Jablonski KA, McAteer JB, De Bakker PIW, et al. Common gene polymorphism on the rosiglitazone response in patients
variants in 40 genes assessed for diabetes incidence and with type 2 diabetes. Diabetes Care 2005;28:1139–44.
PERSONALIZED MEDICINE IN DIABETES MANAGEMENT Elk and Iwuchukwu 1149
53. Morris AD, Boyle DI, MacAlpine R, et al. The diabetes audit 70. Aquilante CL, Zhang W, McCollum M. Race, ethnicity, and
and research in Tayside Scotland (DARTS) study: electronic use of thiazolidinediones among US adults with diabetes. Curr
record linkage to create a diabetes register. DARTS/MEMO Med Res Opin 2007;23:489–94.
Collaboration. BMJ 1997;315:524–8. 71. Aquilante CL, Bushman LR, Knutsen SD, Burt LE, Rome LC,
54. Shu Y, Brown C, Castro RA, et al. Effect of genetic variation Kosmiski LA. Influence of SLCO1B1 and CYP2C8 gene poly-
in the organic cation transporter 1, OCT1, on metformin phar- morphisms on rosiglitazone pharmacokinetics in healthy vol-
macokinetics. Clin Pharmacol Ther 2008;83:273–80. unteers. Hum Genomics 2008;3:7–16.
55. American Diabetes Association. 7. Approaches to glycemic 72. Kirchheiner J, Thomas S, Bauer S, et al. Pharmacokinetics
treatment. Diabetes Care 2016;39(Suppl 1):S52–9. and pharmacodynamics of rosiglitazone in relation to CYP2C8
56. DeFronzo RA. Pharmacologic therapy for type 2 diabetes mel- genotype. Clin Pharmacol Ther 2006;80:657–67.
litus. Ann Intern Med 1999;131:281–303. 73. Gouda HN, Sagoo GS, Harding A-H, Yates J, Sandhu MS, Hig-
57. Aquilante CL. Sulfonylurea pharmacogenomics in type 2 dia- gins JPT. The association between the peroxisome prolifera-
betes: the influence of drug target and diabetes risk polymor- tor-activated receptor-gamma2 (PPARG2) Pro12Ala gene
phisms. Expert Rev Cardiovasc Ther 2010;8:359–72. variant and type 2 diabetes mellitus: a HuGE review and meta-
58. Kirchheiner J, Roots I, Goldammer M, Rosenkranz B, Brock- analysis. Am J Epidemiol 2010;171:645–55.
moller J. Effect of genetic polymorphisms in cytochrome p450 74. Aquilante CL. Pharmacogenetics of thiazolidinedione therapy.
(CYP) 2C9 and CYP2C8 on the pharmacokinetics of oral Pharmacogenomics 2007;8:917–31.
antidiabetic drugs: clinical relevance. Clin Pharmacokinet 75. Bluher M, Lubben G, Paschke R. Analysis of the relationship
2005;44:1209–25. between the Pro12Ala variant in the PPAR-gamma2 gene and
59. Group UKPDS. Intensive blood-glucose control with sulpho- the response rate to therapy with pioglitazone in patients with
nylureas or insulin compared with conventional treatment and type 2 diabetes. Diabetes Care 2003;26:825–31.
risk of complications in patients with type 2 diabetes. Lancet 76. Hsieh MC, Lin KD, Tien KJ, et al. Common polymorphisms
1998;352:837–53. of the peroxisome proliferator-activated receptor-gamma
60. Schopman J, Simon A, Hoefnagel S, Hoekstra J, Scholten R, (Pro12Ala) and peroxisome proliferator-activated receptor-
Holleman F. The incidence of mild and severe hypoglycaemia gamma coactivator-1 (Gly482Ser) and the response to pioglita-
in patients with type 2 diabetes mellitus treated with sulfony- zone in Chinese patients with type 2 diabetes mellitus. Meta-
lureas: a systematic review and meta-analysis. Diabetes Metab bolism 2010;59:1139–44.
Res Rev 2014;30:11–22. 77. Kang ES, Cha BS, Kim HJ, et al. The 11482G>A polymor-
61. Li Q, Chen M, Zhang R, et al. KCNJ11 E23K variant is associ- phism in the perilipin gene is associated with weight gain with
ated with the therapeutic effect of sulphonylureas in Chinese rosiglitazone treatment in type 2 diabetes. Diabetes Care
type 2 diabetic patients. Clin Exp Pharmacol Physiol 2006;29:1320–4.
2014;41:748–54. 78. Bailey SD, Xie C, Do R, et al. Variation at the NFATC2 locus
62. Hamming KSC, Soliman D, Matemisz LC, et al. Coexpression increases the risk of thiazolidinedione-induced edema in the
of the type 2 diabetes susceptibility gene variants KCNJ11 Diabetes REduction Assessment with ramipril and rosiglita-
E23K and ABCC8 S1369A alter the ATP and sulfonylurea sen- zone Medication (DREAM) study. Diabetes Care 2010;33:
sitivities of the ATP-sensitive K+ channel. Diabetes 2250–3.
2009;58:2419–24. 79. Drucker DJ, Nauck MA. The incretin system: glucagon-like
63. Javorsky M, Klimcakova L, Schroner Z, et al. KCNJ11 gene peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibi-
E23K variant and therapeutic response to sulfonylureas. Eur J tors in type 2 diabetes. Lancet 2006;368:1696–705.
Intern Med 2012;23:245–9. 80. Kleinberger JW, Pollin TI. Personalized medicine in diabetes
64. Sesti G, Laratta E, Cardellini M, et al. The E23K Variant of mellitus: current opportunities and future prospects. Ann N Y
KCNJ11 encoding the pancreatic b-cell adenosine 50 -tripho- Acad Sci 2015;1346:45–56.
sphate-sensitive potassium channel subunit Kir6.2 is associated 81. ’t Hart LM, Fritsche A, Nijpels G, et al. The CTRB1/2 locus
with an increased risk of secondary failure to sulfonylurea in affects diabetes susceptibility and treatment via the incretin
patients with type 2 diabetes. J Clin Endocrinol Metab pathway. Diabetes 2013;62:3275–81.
2006;91:2334–9. 82. Nathan DM, Buse JB, Kahn SE, et al. Rationale and design
65. Kimber CH, Doney AS, Pearson ER, et al. TCF7L2 in the Go- of the glycemia reduction approaches in diabetes: a compara-
DARTS study: evidence for a gene dose effect on both diabetes tive effectiveness study (GRADE). Diabetes Care 2013;36:
susceptibility and control of glucose levels. Diabetologia 2254–61.
2007;50:1186–91. 83. Hivert M-F, Jablonski KA, Perreault L, et al. Updated genetic
66. Sesti G, Marini MA, Cardellini M, et al. The Arg972 variant score based on 34 confirmed type 2 diabetes loci is associated
in insulin receptor substrate-1 is associated with an increased with diabetes incidence and regression to normoglycemia in
risk of secondary failure to sulfonylurea in patients with type the diabetes prevention program. Diabetes 2011;60:1340–8.
2 diabetes. Diabetes Care 2004;27:1394–8. 84. Meigs JB, Shrader P, Sullivan LM, et al. Genotype score in
67. Prudente S, Morini E, Lucchesi D, et al. IRS1 G972R missense addition to common risk factors for prediction of type 2 dia-
polymorphism is associated with failure to oral antidiabetes betes. N Engl J Med 2008;359:2208–19.
drugs in white patients with type 2 diabetes from Italy. Dia- 85. Diabetes Genes. Genetic types of diabetes including maturity-
betes 2014;63:3135–40. onset diabetes of the young (MODY). Available from www.di
68. van Leeuwen N, Swen JJ, Guchelaar HJ, ’t Hart LM. The role abetesgenes.org. Accessed February 13, 2017.
of pharmacogenetics in drug disposition and response of oral 86. Genetic Testing Registry. Available from www.ncbi.nlm.nih.
glucose-lowering drugs. Clin Pharmacokinet 2013;52:833–54. gov/gtr/. Accessed February 13, 2017.
69. Della-Morte D, Palmirotta R, Rehni AK, et al. Pharmacoge- 87. Monogenic Diabetes Registry. Available from http://monogenic
nomics and pharmacogenetics of thiazolidinediones: role in diabetes.uchicago.edu/mody-registry/. Accessed February 13,
diabetes and cardiovascular risk factors. Pharmacogenomics 2017.
2014;15:2063–82.

You might also like