PHARMACOGENETICS IN DIABETES - EWAN R. PEARSON, MBBCHIR, PHD

Page created by Cory Bowers
 
CONTINUE READING
Pharmacogenetics in Diabetes
Ewan R. Pearson, MBBChir, PhD

Corresponding author                                                 The response to a drug is determined by the con-
Ewan R. Pearson, MBBChir, PhD                                    centration of active drug available at its site(s) of action
Biomedical Research Institute, Ninewells Hospital and Medical    (pharmacokinetics) and the ability of the drug to elicit an
School, Ninewells Avenue, Dundee, DD1 9SY, United Kingdom.
E-mail: e.pearson@chs.dundee.ac.uk
                                                                 effect at its site of action (pharmacodynamics). In phar-
                                                                 macokinetics, drugs can be actively transported across
Current Diabetes Reports 2009, 9:172–181
Current Medicine Group LLC ISSN 1534-4827                        the gut epithelium, into the target tissue, or into the
Copyright © 2009 by Current Medicine Group LLC                   renal tubules. In addition, drugs are often metabolized
                                                                 to an active form, or are metabolized to an inactive form
                                                                 before excretion from the body. Historically, pharmaco-
  Genetic variation can impact on efficacy and risk              genetics has focused on the study of variation in the drug
  of adverse events to commonly used oral agents in              metabolizing enzymes, in particular the cytochrome P450
  diabetes. Metformin is not metabolized and its mech-           enzymes. However, recent developments have occurred in
  anism of action remains debated; however, several              the understanding of drug transport with some exciting
  cation transporters have been identified. Variation in         new data on how variation in genes encoding transporters
  these pharmacokinetic genes might influence metfor-            might impact on drug response.
  min response. Conversely, although the cytochrome                  The pharmacodynamics of a drug can be broadly
  P450 system has been implicated in sulfonylurea                divided into direct and indirect factors. The direct drug
  response in some small studies, to date variants affect-       effects will be influenced by its ability to bind to a receptor,
  ing pharmacodynamics, including those in ABCC8                 the function of that receptor, and the function of the down-
  (SUR1) and TCF7L2, are the most promising. For                 stream pathways. The indirect factors are those that are
  thiazolidinedione response, variants in PPARG or               distinct from the effector pathway (eg, response to a drug
  ADIPOQ (adiponectin) have been variably associ-                that increases insulin secretion may well be more effective
  ated with response. With increasing well-phenotyped            in a patient who is more insulin sensitive, although the
  cohorts and new methods, including genome-wide                 drug effect has no effect on insulin action). For a disease
  association studies, the next few years offer great hope       such as type 2 diabetes (which is highly heterogeneous,
  to use pharmacogenetics to unravel drug and disease            and in which the drugs used target the disease-causing
  mechanisms, as well as the possibility to individualize        defects), direct and indirect pharmacodynamics of a drug
  therapy by genotype.                                           will be influenced by disease etiology (ie, an individual
                                                                 may respond well to sulfonylureas because their diabetes
                                                                 is etiologically distinct from an individual who responds
Introduction                                                     poorly). Therefore, it may be possible to use drug response
Pharmacogenetics is the study of how genetic variation           rather than the traditional case/control study, as a tool to
affects drug response—either drug efficacy or adverse out-       investigate diabetes etiology.
come. Although in the treatment of diabetes, hypertension
and hyperlipidemia are also targeted, this article focuses on
the pharmacogenetics of oral antihyperglycemic medica-           Pharmacokinetic Pharmacogenetics
tion in type 2 diabetes and monogenic diabetes and will not      Sulfonylureas
address insulin treatment or type 1 diabetes. Compared with      In 1979, a ninefold variation in the rate of tolbutamide
anticancer therapy, the field of pharmacogenetics of diabetes    disappearance from plasma was described with a trimodal
is in its infancy. However, genetics has impacted on diabetes    distribution suggestive of monogenic inheritance [1]. This
treatment in some clear areas. With the exponential growth       variation in hydroxylation of tolbutamide was subsequently
of large-scale genotyping methods, including genome-wide         shown to be due to variation in CYP2C9 [2]. CYP2C9
association (GWA) study, we may be able to identify further      has also been shown to be a rate-limiting enzyme in the
variants that impact on response or side effects. In addition,   metabolism of other sulfonylureas, including glibenclamide
as will be discussed, drug response potentially can be used      [3], gliclazide [4], glipizide [5], and glimepiride [6]. Two
to investigate drug mechanisms and disease etiology.             variants in CYP2C9 affect the catalytic function of the
Pharmacogenetics in Diabetes
                                                                                                              I   Pearson
                                                                                                                            I   173

enzyme: Arg144Cys (2C9*2; allele frequency 11%) and Ile-       required to determine the clinical impact of variation in
359Leu (2C9*3; allele frequency 7%). For glibenclamide,        this drug transporter in the diabetic population.
the clearance for the *2/*2 individuals was reduced by
25%, and for the *3/*3 individuals by 57% compared             Thiazolidinediones
with wild-type [3]. Similar figures for tolbutamide are        Thiazolidinediones are extensively metabolized in the
25% and 84% [7].                                               liver, predominantly by CYP2C8, with CYP2C9 (pio-
    Despite the clear data showing dramatic differences in     glitazone and rosiglitazone) and CYP3A4 (pioglitazone)
sulfonylurea metabolism for carriers of different CYP2C9       playing a minor role. The data on the role of CYP2C8
variants, minimal study has been done on the effect of         on thiazolidinedione response are inconclusive. The in
these variants on response (Table 1). One recent study sug-    vitro work suggests that the 2C8*3 variant should impair
gests, for the first time, that CYP2C9 variants may impact     metabolism, yet in response to rosiglitazone, carriers of
on sulfonylurea prescribing. In a retrospective observa-       the 2C8*3 polymorphism (Arg139Lys and Lys399Arg
tional study of 296 patients prescribed tolbutamide, those     substitutions) had lower elimination half-lives than wild-
carrying the *3 allele of CYP2C9 did not have their tolbu-     type but showed no difference in glucose lowering [16].
tamide dose increased when compared with the wild-type         For pioglitazone, 2C9*3 polymorphisms reduced the area
(*1/*1) group [8]. This effect was not seen in a smaller       under the plasma concentration time curve [17]; however,
number of patients treated with glibenclamide (n = 76)         no studies have looked at the effect of this genotype on
and glimepiride (n = 76). This could reflect lack of power.    pharmacodynamic response.
However, it may be important to look at other cytochrome
P450 genes because a study in a Chinese population recently
showed that gliclazide modified release is predominantly       Pharmacodynamic Pharmacogenetics
metabolized by CYP2C19 rather than 2C9 [9].                    Sulfonylureas
                                                               Sulfonylureas bind to the SUR1 moiety of the pancreatic
Metformin                                                      β-cell K ATP channel causing the channel to close and trig-
Metformin is not metabolized and is primarily excreted         ger insulin secretion. The fact that genetic variation in this
unchanged in the urine. Recent studies have implicated         pathway can alter sulfonylurea response is best highlighted
the role of organic cation transporters in metformin           by the 1% to 2% of diabetes caused by Mendelian muta-
disposition: PMAT (plasma membrane monoamine trans-            tions in TCF1 (encoding hepatocyte nuclear factor-1 α
porter) (SLC29A4) is involved in gut absorption [10],          [HNF-1α]) causing maturity-onset diabetes of the young.
OCT1 (SLC22A1) primarily in hepatic uptake, and OCT2           In a randomized trial of sulfonylureas and metformin in
(SLC22A2) in tubular secretion [11,12].                        patients with diabetes due to TCF1 mutations and type
     In a transgenic mouse model, knockout of liver            2 diabetes, the fall in fasting plasma glucose (FPG) into
Slc22a1 virtually abolished hepatic lactate production         gliclazide was 3.9-fold greater in patients with TCF1
supporting a key role of Oct1 in transporting metfor-          mutations than their response to metformin (P = 0.002);
min into the hepatocytes [13]. An elegant study by Shu         as expected, no difference in response to gliclazide or met-
et al. [14••] took this concept further and showed that        formin was apparent in those with type 2 diabetes [18].
OCT1 plays an important role in determining metfor-            The mechanism probably results from the fact that the
min response in humans. They showed that deletion              major β-cell defects due to reduced HNF-1α function are
of Slc22a1 in mouse liver reduced metformin effects on         in glucose metabolism, and are therefore bypassed by sul-
5′ adenosine monophosphate-activated protein kinase            fonylureas that act on the K ATP channel to stimulate insulin
(AMPK) phosphorylation and gluconeogenesis; as a con-          release [18]. This study highlighted, for the fi rst time, the
sequence, the glucose-lowering effect of metformin was         importance of genetic etiology in determining response
abolished. In addition, they described four loss-of-func-      to treatment in diabetes and has led to change in clinical
tion polymorphisms in SLC22A1 that in a study of 20            management of patients with TCF1 mutations. Sulfonyl-
normal glucose-tolerant individuals reduced the effect of      ureas are now recommended as the fi rst-line antidiabetic
metformin on response to oral glucose [14••]. In a subse-      therapy for these patients. It is exciting that patients who
quent paper, they demonstrated higher serum metformin          have been assumed to have type 1 diabetes and are treated
concentrations in those carrying the reduced function          with insulin, who are subsequently found to have a TCF1
OCT1 polymorphisms, suggesting that this is due to             mutation, have been able to transfer off insulin onto sulfo-
reduced hepatic uptake of the drug [15]. In contrast to the    nylurea therapy [19].
fi ndings by Shu et al. [14••], a study of 24 responders and       A further example of how identifying monogenic dia-
nine non-responders to metformin showed no difference          betes can impact dramatically on diabetes treatment can be
in the prevalence of OCT1 or OCT2 variants between             seen in the recent discoveries in neonatal diabetes. In 2004,
these two groups. A large study of SLC22A1 variation           a third of cases of diabetes diagnosed before 6 months of
on glycemic response and side effects to metformin is          age were found to be due to activating mutations in the
174

Table 1. Pharmacogenetic studies on sulfonylurea outcome assessed by clinical response with more than 1 month of oral treatment
                                                                                                                                                                                  I

Study                  Study population                         Intervention and outcome                 Gene polymorphisms           Outcome
Pharmacokinetic
                                                                                                                                                                                  Genetics

Holstein et al. [55]   Any sulfonylurea, 20 patients with       Admissions to emergency department CYP2C9 *2 and *3                   2 of the hypoglycemic group (10%)
                        severe hypoglycemia vs 337 without       with hypoglycemia                                                     were *2/*3 or *3/*3 compared with 7
                                                                                                                                       (2.1%) of the controls
Becker et al. [8]      Observational study, 248 patients       Change in daily prescribed dose           CYP2C9 *2 and *3             Glibenclamide NS, tolbutamide *1/*3
                        treated with sulfonylurea for at least                                                                         and *2/*3 12-mg dose increment
                        10 prescriptions (172 tolbutamide,                                                                             compared with 279-mg dose incre-
                        42 glimepiride, 34 glibenclamide)                                                                              ment for wild-type (*1/*1) (P = 0.009);
                                                                                                                                       glimepiride NS
Pharmacodynamics
Gloyn et al. [29]      Randomized prospective study             Reduction in fasting glucose at 1 y,     KCNJ11 E23K and L270V        No significant genotypic effect
                        (UKPDS), 363 patients treated with       or sooner if failed on sulfonylurea
                        chlopropamide, glibenclamide,            treatment before 1 y
                        or glipizide
Sesti et al. [28]      Prospective study, 525 patients treated Binary outcome–sulfonylurea failure KCNJ11 E23K                        Carriers of K allele (RR for treatment
                        with glibenclamide until failure when are those requiring insulin after                                        failure, 1.45; P = 0.04) compared
                        metformin was added                     failure of combined sulfonylurea and                                   with wild-type
                                                                metformin treatment
Sesti et al. [56]      As above                                 As above                                 Gly972Arg IRS-1              Carriers of Arg972 IRS-1 variant
                                                                                                                                       associated with treatment failure (RR,
                                                                                                                                       2.7 [1.02–7.28]; P = 0.045) compared
                                                                                                                                       with wild-type
Feng et al. [26••]     Prospective study, gliclazide for 8 wk; FPG reduction at 6 wk; HbA1c reduction Ser1369Ala of ABCC8             In the combined group, subjects with
                        initial round (n = 661), replication                                                                            Ala/Ala had a 7.7% greater FPG
                        (n = 607)                                                                                                       reduction (P < 0.001), an 11.9% greater
                                                                                                                                        decrease in 2-hour plasma glucose
                                                                                                                                        (P = 0.003) compared with Ser/Ser. No
                                                                                                                                        difference in HbA1c reduction
FPG—fasting plasma glucose; HbA1c—hemoglobin A1c; NS—not significant; RR—relative risk; UKPDS—United Kingdom Prospective Diabetes Study.
Pharmacogenetics in Diabetes
                                                                                                              I   Pearson
                                                                                                                            I   175

KCNJ11 gene encoding the Kir6.2 subunit of the β-cell            as failure of combination sulfonylurea and metformin
K ATP channel [20]. Subsequently, in 2006, mutations in          therapy rather than sulfonylurea alone. In this study,
the ABCC8 gene encoding the other subunit of the K ATP           carriers of the K allele had a relative risk for failure of
channel, SUR1, were also found to cause neonatal diabetes,       this combination of 1.45 (95% CI, 1.01–2.09; P = 0.04).
although less commonly [21,22]. With these mutations,            However, it is unclear whether this reflects sulfonylurea
the pancreatic K ATP channel is insensitive to the increase in   failure, metformin failure, or simple differential rates in
intracellular adenosine triphosphate/adenosine diphosphate       diabetes progression by genotype.
that results from glucose metabolism. Thus, the pancreatic           In 2006, the DECODE group published an associa-
β cell does not secrete insulin in response to hyperglycemia.    tion between TCF7L2 variants and type 2 diabetes risk,
Sulfonylureas bind to the K ATP channel, and intravenous         such that the 10% of the population homozygous for
tolbutamide was able to stimulate insulin secretion in           the risk variant were twice as likely to develop diabetes
patients with KCNJ11 mutations [20]. This work led to the        as the wild-type population [30•]. This has been widely
successful transfer of patients with neonatal diabetes who       replicated and remains the strongest genetic association
had lifelong insulin treatment to oral sulfonylurea therapy      for type 2 diabetes described to date. The mechanism for
with near normalization of blood glucose [23,24].                how TCF7L2 variants cause diabetes remains unclear,
    Can this monogenic paradigm be applied to common             although several studies point to this being due to
type 2 diabetes? Do polymorphisms in glucose metabolism          decreased β-cell function [31–33], possibly mediated by
enzymes, the K ATP channel, or downstream pathways influ-        an impaired incretin response [32]. Given the potential
ence sulfonylurea response? Recently, several established        role of TCF7L2 in insulin secretion, and its large effect
variants have been identified as associated with type 2 dia-     (by type 2 diabetes standards), it is a good candidate
betes risk that impact primarily on β-cell insulin secretion;    gene for assessing impact on sulfonylurea response. In
these include variants in TCF7L2, KCNJ11, CDKAL1,                a large study from Tayside, Scotland, of 901 incident
CDKN2A-2B, WFS1, HHEX-IDE, and SLC30A8 [25]. To                  users of sulfonylureas, patients with type 2 diabetes who
date, the only etiologic candidate genes published investigat-   were homozygous for the diabetes risk allele (G) at SNP
ing association with sulfonylurea response are in KCNJ11,        rs12255372 were twice as likely not to be treated to below
ABCC8, and TCF7L2. Once again, the field is relatively           a target HbA1c of 7% in the fi rst 3 to 12 months of treat-
small and the results are conflicting (Table 1). However,        ment compared with patients homozygous for the T allele
a few reasonable sized studies of interest exist. In what is     (OR, 1.95; P = 0.005) [34•]. Importantly, no effect was
probably the largest and cleanest diabetes pharmacogenetics      observed of this variant on metformin response (n = 945)
study to date, 25 single nucleotide polymorphisms (SNPs) in      [34•], showing that the association is with sulfonylurea
11 candidate genes were examined in a prospective trial of       response rather than diabetes severity or progression.
1268 patients treated with gliclazide. After an initial round
(n = 661), the Ser1369Ala of the ABCC8 gene and rs5210           Metformin
of the KCNJ11 gene were significantly associated with            As detailed above, the main success in metformin phar-
decreases in FPG (P = 0.002). This finding for Ser1369Ala        macogenetics has been the elucidation of different drug
was replicated in a separate cohort (n = 607). In the com-       transporters. The picture is not as clear for pharmacody-
bined groups, compared with subjects with the Ser/Ser            namic pharmacogenetics as, in contrast to sulfonylureas,
genotype, subjects with the Ala/Ala genotype had a 7.7%          the exact mechanistic pathway for metformin remains
greater decrease in FPG (P < 0.001), and an 11.9% greater        unclear. At a physiologic level, metformin’s primary effect
decrease in 2-hour plasma glucose (P = 0.003), although no       is probably to reduce hepatic glucose output by increasing
difference in hemoglobin A1c (HbA1c) was seen [26••].            insulin suppression of gluconeogenesis [35•]. However,
    KCNJ11 (encoding the Kir6.2 subunit of the K ATP             debate exists over its role in augmenting insulin-mediated
channel) is adjacent to ABCC8 on chromosome 11, and              glucose disposal into muscle, and an often overlooked
ABCC8 Ser1369Ala and KCNJ11 rs5210 and E23K are in               mechanism of metformin is a reduction in noninsulin-
strong linkage disequilibrium (ie, highly correlated). The       mediated glucose clearance [35•], which explains as much
E23K variant of KCNJ11 was robustly associated with              of the effect of metformin on glucose lowering as its role
type 2 diabetes in a large meta-analysis [27]. In a study        on hepatic glucose production, and a reduction in glucose
of human donor islets, glibenclamide-induced insulin             absorption from the gut.
secretion was impaired in the KK islets [28]. The associa-           At a molecular level, metformin’s effects are mediated
tion of response to sulfonylureas and the E23K variant           via AMPK, an effect that requires phosphorylation of
were studied in the UKPDS cohort, where in a study of            AMPK by LKB1, but metformin does not directly activate
360 type 2 diabetic patients, no effect of the genotype          AMPK or LKB1, and the mechanism by which metfor-
occurred on the change in FPG in the fi rst year of treat-       min activates AMPK remains to be determined. This
ment [29]. In a subsequent study of 525 patients with type       physiologic and molecular uncertainty makes it difficult
2 diabetes, sulfonylurea failure was confusingly defi ned        to hypothesize good candidate genes. However, obvious
176

Table 2. Pharmacogenetic studies on thiazolidinedione outcome assessed by clinical response with more than 1 month of oral treatment
                                                                                                                                                                                 I

Study                         Study population and intervention Outcome                                 Gene polymorphisms                   Outcome
Pharmacokinetic
                                                                                                                                                                                 Genetics

None
Pharmacodynamic PPARG
Bluher et al. [43]            Prospective study of 131 patients Reduction in HbA1c or FPG at 12 PPARG Pro12Ala                               No significant genotypic effect
                               with type 2 diabetes treated with and 26 wk
                               pioglitazone, 45 mg, for > 26 wk
Snitker et al. [44]           93 women with previous gestational Change in insulin sensitivity at       PPARG Pro12Ala                       No significant genotypic effect
                               diabetes treated with troglitazone, 12 wk determined by IVGTT.
                               400 mg, daily for 12 wk             Nonresponders (lower tertile)
                                                                   compared with responders
                                                                   (upper 2 tertiles)
Wolford et al. [45]           As above                              As above                            PPARG sequenced, identifying 61 8 SNPs associated with response
                                                                                                         SNPs (MAF > 5%). Single SNP     (none significant after adjusting
                                                                                                         and haplotype analysis          for multiple testing); 3 of these
                                                                                                                                         SNPs (rs4135263, rs10510419,
                                                                                                                                         rs1152003) were marginally asso-
                                                                                                                                         ciated with changes in insulin
                                                                                                                                         sensitivity (as quantitative trait);
                                                                                                                                         3 haplotypes were marginally
                                                                                                                                         associated with response
Kang et al. [47]              Prospective study, 198 patients       FPG and HbA1c reduction at 3 mo PPARG Pro12Ala; NB Ala          Decrease in FPG (3.20 mmol/L vs
                               treated with rosiglitazone, 4 mg,                                     frequency very low in this      1.35; P = 0.003) and decrease in
                               for 3 mo                                                              Korean population (MAF 3%) so HbA1c (1.66 vs 0.48; P = 0.012)
                                                                                                     only 11 Pro/Ala and no Ala/Ala  greater in Ala carriers compared
                                                                                                     individuals                     with wild-type
Florez et al. [46]            Prospective study, 340 nondiabetic Change in insulin sensitivity          PPARG Pro12Ala, and 5                No significant genotypic effect on
                               subjects with IGT or IFG           index derived from OGTT                SNPs from Wolford et al.             FPG or HbA1c reduction
                                                                                                         [45] (rs880663, rs4135263
                                                                                                         rs1152003, rs6806708, and
                                                                                                         rs13065455)
Pharmacodynamic other
Kang et al. [48]              Prospective study, rosiglitazone,     FPG and HbA1c reduction at 6 wk ADIPOQ (adiponectin) SNP                 Patients G/G at +45 or +276 had
                               4 mg, every day for 3 mo                                              +45T/G and SNP +276G/T                   smaller FPG (P = 0.032 and
                                                                                                                                              P = 0.001) and HbA1c (P = 0.028
                                                                                                                                              and P = 0.006) reduction
                                                                                                                                              with rosiglitazone
FPG—fasting plasma glucose; HbA1c—hemoglobin A1c; IFG—impaired fasting glucose; IGT—impaired glucose tolerance; IVGTT—intravenous glucose tolerance test; MAF—minor allele
frequency; OGTT—oral glucose tolerance test; SNP—single nucleotide polymorphism.
Table 2. Pharmacogenetic studies on thiazolidinedione outcome assessed by clinical response with more than 1 month of oral treatment                              (Continued)

Study                         Study population and intervention Outcome                                 Gene polymorphisms                   Outcome
Sun et al. [49]               42 patients treated with 4 mg of      FPG reduction                       ADIPOQ; -11377 C/G, +45 T/G          No significant genotypic effect of
                               rosiglitazone daily for 12 wk                                                                                  +45T/G. Lower FPG reduction
                                                                                                                                              in carriers of G allele at -11377
                                                                                                                                              (although baseline significantly
                                                                                                                                              lower in this group)
Kang et al. [57]              Rosiglitazone, 4 mg, daily for 3 mo FPG and HbA1c reduction               Perilipin; 6209 G/A, 11482 G/A,      No significant genotypic effect on
                                                                                                         13041 A/G, 14995 A/T                 FPG or HbA1c reduction
Wang et al. [58]              113 patients treated with             > 10% reduction in FPG or        Lipoprotein lipase; S447X               S/S genotype associated with
                               pioglitazone, 30 mg, for 10 wk        absolute reduction in HbA1c >1%                                          increased response
Wang et al. [51]              93 patients treated with 4 or 8 mg > 15% FPG reduction or absolute ABCA1; R219K, M883I,                        For R219K, KK had more treatment
                               of rosiglitazone for 48 wk         reduction in HbA1c > 0.5%       and R1587K                                  failures than relative risk (per allele
                                                                                                                                              OR, 2.14; P < 0.05; not adjusted
                                                                                                                                              for multiple comparisons). No
                                                                                                                                              significant genotypic effect of
                                                                                                                                              M883I or R1587K
FPG—fasting plasma glucose; HbA1c—hemoglobin A1c; IFG—impaired fasting glucose; IGT—impaired glucose tolerance; IVGTT—intravenous glucose tolerance test; MAF—minor allele
frequency; OGTT—oral glucose tolerance test; SNP—single nucleotide polymorphism.
                                                                                                                                                                                        Pharmacogenetics in Diabetes
                                                                                                                                                                                        I
                                                                                                                                                                                        Pearson
                                                                                                                                                                                        I
                                                                                                                                                                                        177
178
      I   Genetics

candidates are the genes encoding the AMPK subunits. A         patients had no beneficial effect of troglitazone on insulin
Japanese study of 192 cases and 272 controls identified        sensitivity. An initial analysis showed no effect of the Pro-
a haplotype across PRKAA2 (encoding the AMPK-α2                12Ala polymorphism on this response [44]; however, in a
subunit) that was associated with type 2 diabetes, was         subsequent detailed haplotype analysis across the PPARG
replicated in two independent cohorts, and was associ-         gene, a weak association with response was found with
ated with insulin resistance assessed by homeostasis           certain haplotypes, although the small numbers here
model assessment [36]. However, a haplotype analysis           make this haplotype analysis underpowered [45]. Florez
in 4206 Scandinavian and Canadian individuals showed           et al. [46] also studied the influence of PPARG variants
no association between PRKAB1 (encoding the AMPK-              on the effect of troglitazone on insulin sensitivity index
β1 subunit) and PRKAB2 (encoding AMPK-β2 subunit)              in the Diabetes Prevention Program but showed no
and type 2 diabetes risk or insulin sensitivity [37]. Fur-     association with response. The only result suggesting an
thermore, a study of 1787 unrelated Japanese subjects          association with response was in a Korean population of
found no association between PRKAA2, STK11 (LKB1),             198 patients treated with rosiglitazone, 4 mg, daily. They
and CRTC2 (TORC2) variants and type 2 diabetes that            reported that those carrying the Ala allele had a greater
withstood correction for multiple testing [38]. Thus, no       response to rosiglitazone than the Pro/Pro homozygotes;
convincing evidence exists for an association between the      however, the allele frequency in the Ala group was very
pathways of metformin action and diabetes risk. However,       low (3%), and this result is only based on 11 Pro/Ala and
just because variants in this pathway do not explain dia-      no Ala/Ala individuals [47].
betes risk does not preclude them from having an effect on         Genes other than PPARG have been studied for asso-
metformin response, and the haplotypes identified would        ciation with thiazolidinedione response (Table 2). A couple
be good candidates to assess metformin response. Inter-        of groups looked at the adiponectin gene ADOPOQ,
estingly, in a small study in which metformin was used         but again with no consistent replicated results [48,49].
to induce ovulation in patients with polycystic ovarian        An interesting alternative candidate came from a mouse
syndrome, variation in rs8111699 of STK11 (LKB1) was           model lacking the high-density lipoprotein synthesis gene
significantly associated with ovulation induction success:     Abca1. Mice lacking Abca1 exhibited lipid accumulation
48% (10/21) of C/C women, 67% (32/48) of C/G women,            and β-cell toxicity [50]. Rosiglitazone activates Abca1 and
and 79% (15/19) of G/G women ovulated [39]. Although           therefore could protect against this β-cell lipotoxicity. In
the study numbers were very small, it would be interesting     just one study, one SNP was identified in ABCA1 that was
to look at this variant with respect to glycemia or insulin    nominally associated with response to rosiglitazone [51].
sensitivity outcomes.                                              Thiazolidinediones are associated with increased risk of
                                                               heart failure and fluid retention. This has been attributed to
Thiazolidinediones                                             PPAR-γ regulation of a renal-collecting duct sodium trans-
Thiazolidinediones promote the binding of the transcrip-       porter (ENAC), and polymorphisms in the gene encoding
tion factor peroxisome proliferator-activated receptor-γ       the ENAC β subunit have been associated significantly
(PPAR-γ) to its DNA response element. Among several            with edema in a study of 207 patients receiving farglitazar
effects, the thiazolidinediones promote adipocyte dif-         in phase 3 clinical trials [52]. In a study of another glitazar
ferentiation. Physiologically, thiazolidinediones increase     (dual-acting PPAR-γ/-α agonists), ragaglitazar, edema was
insulin-stimulated glucose uptake into muscle, insulin         less in those carrying the protective Ala allele at Pro12Ala
suppression of hepatic glucose output, and insulin-stimu-      PPAR-γ than the wild-type patients, and was not influenced
lated lipolysis [40].                                          by the Leu162Val SNP in PPAR-α [53].
    Variation at PPARG (encoding PPAR-γ) was one of
the fi rst loci to be robustly associated with type 2 dia-
betes, with the fi nding that carriers of the Ala variant at   Future Directions for Diabetes Pharmacogenetics
codon 12 were protected against diabetes with a per-allele     The arrival of the GWA study has had a dramatic impact
RR of 1.25 compared with the Pro/Pro individuals [41].         on gene discovery for disease traits, including type 2 dia-
Because the Pro12Ala variant influences transcriptional        betes. The great advantage of these studies is they make
activity of PPARG and is located in the ligand-binding         no prior assumption about mechanism and, as such, their
domain, this variant is a good candidate to affect thiazoli-   primary role in diabetes gene discovery has been to reveal
dinedione response [42]. Several groups have studied this      novel pathways not previously thought to be associated
and found variable results, probably reflecting the small      with diabetes. Therefore, if drug response (efficacy or
sample sizes in each group (Table 2). Bluher et al. [43]       adverse outcome) is used as the outcome of interest, a GWA
found no association with Pro12Ala and HbA1c reduction         study could reveal new insights into drug mechanism. For
with pioglitazone, 45 mg, in 131 patients. In an analysis      example, this might help unravel the mechanism of action
of the TRIPOD study, which treated women with pre-             of metformin, or the mechanism of gastrointestinal intol-
vious gestation diabetes with troglitazone, one third of       erance in up to 25% of individuals who are treated with
Pharmacogenetics in Diabetes
                                                                                                                I   Pearson
                                                                                                                              I   179

metformin. In addition, because disease etiology is an         Clinical Trial Acronyms
important determinant of drug response, a GWA on drug          DECODE—Diabetes Epidemiology: Collaborative analysis
response could reveal new etiologic variants. However,         of Diagnostic criteria in Europe; TRIPOD—Troglitazone
a drawback for this approach is that large sample sizes        in Prevention of Diabetes; UKPDS—United Kingdom Pro-
and independent replication cohorts are required. At the       spective Diabetes Study.
present time, pharmacogenetic studies are too small and
rarely replicated, so efforts must be made to collect better
cohorts and form collaborations to increase sample sizes.      Acknowledgments
Another limitation of the GWA approach is that it is only      Ewan R. Pearson holds a clinician scientist fellowship (CSO/
able to detect common variation. It is likely that genetic     NHS Scotland), and his work is supported by Diabetes UK
effects that will impact enough on drug response to affect     (grant 07/0003525).
clinical prescribing will be rare, and therefore missed by
a GWA study. However, a recent study on myopathy in
statin users provided great support for using the GWA          Disclosure
approach in pharmacogenetics. In this study, the genetic       No potential confl ict of interest relevant to this article
effect size was so large that it was possible to perform a     was reported.
genome-wide scan on just 85 patients with severe myopa-
thy and 90 controls on high-dose statins [54••]. This
contrasts with the sample sizes of greater than 30,000         References and Recommended Reading
that are now being studied in meta-analyses of multiple        Papers of particular interest, published recently,
GWA studies to fi nd disease association with a relative       have been highlighted as:
risk of 1.1. A “hit” achieving genome-wide significance at     •     Of importance
rs4149056 in SLCO1B1 (encoding OATP1B, an organic              ••    Of major importance
anion transporter involved in statin transport into the
liver) was replicated in a second cohort [54••]. For seri-     1.    Scott J, Poffenbarger PL: Pharmacogenetics of tolbutamide
                                                                     metabolism in humans. Diabetes 1979, 28:41–51.
ous adverse events, which for diabetes drugs might be
                                                               2.    Relling MV, Aoyama T, Gonzalez FJ, Meyer UA: Tolbutamide
heart failure with thiazolidinediones or severe gastro-              and mephenytoin hydroxylation by human cytochrome
intestinal intolerance to metformin, this GWA approach               P450s in the CYP2C subfamily. J Pharmacol Exp Ther 1990,
may help unravel as-yet unknown mechanisms for these                 252:442–447.
                                                               3.    Kirchheiner J, Brockmoller J, Meineke I, et al.: Impact of
adverse events.                                                      CYP2C9 amino acid polymorphisms on glyburide kinetics
                                                                     and on the insulin and glucose response in healthy volunteers.
                                                                     Clin Pharmacol Ther 2002, 71:286–296.
                                                               4.    Elliot DJ, Suharjono, Lewis BC, et al.: Identification of
Conclusions                                                          the human cytochromes P450 catalysing the rate-limiting
In the past few years, considerable advances have been               pathways of gliclazide elimination. Br J Clin Pharmacol
made in our knowledge of genetic determinants of dia-                2007, 64:450–457.
betes and in our knowledge of drug disposition and             5.    Kidd RS, Curry TB, Gallagher S, et al.: Identification of a
                                                                     null allele of CYP2C9 in an African-American exhibiting
action. To date, most pharmacogenetics studies on dia-               toxicity to phenytoin. Pharmacogenetics 2001, 11:803–808.
betes therapies are small and nonreplicated. This in part      6.    Wang R, Chen K, Wen SY, et al.: Pharmacokinetics of
reflects the limited study populations who are suitably              glimepiride and cytochrome P450 2C9 genetic polymor-
                                                                     phisms. Clin Pharmacol Ther 2005, 78:90–92.
phenotyped for drug response, and a clear need exists          7.    Kirchheiner J, Bauer S, Meineke I, et al.: Impact of CYP2C9
for larger cohorts in which drug response is well char-              and CYP2C19 polymorphisms on tolbutamide kinetics
acterized. DNA collection is a surprising omission from              and the insulin and glucose response in healthy volunteers.
                                                                     Pharmacogenetics 2002, 12:101–109.
several large recent pharmaceutical trials on diabetes
                                                               8.    Becker ML, Visser LE, Trienekens PH, et al.: Cytochrome
drugs, so we are currently limited to observational stud-            P450 2C9 *2 and *3 polymorphisms and the dose and effect
ies and small prospective trials. Therefore, it is even more         of sulfonylurea in type II diabetes mellitus. Clin Pharmacol
important to collaborate and replicate results before                Ther 2008, 83:288–292.
                                                               9.    Zhang Y, Si D, Chen X, et al.: Influence of CYP2C9 and
publication to minimize the amount of “false-positives”              CYP2C19 genetic polymorphisms on pharmacokinetics
in the literature. Great potential exists for pharma-                of gliclazide MR in Chinese subjects. Br J Clin Pharmacol
cogenetics to deliver in the next few years. Currently,              2007, 64:67–74.
                                                               10.   Zhou M, Xia L, Wang J: Metformin transport by a newly
the organic cation transporters offer the most hope for              cloned proton-stimulated organic cation transporter
determinants of metformin response and genes affecting               (plasma membrane monoamine transporter) expressed in
β-cell function are the most promising candidates for                human intestine. Drug Metab Dispos 2007, 35:1956–1962.
sulfonylurea response. However, the use of genome-wide         11.   Wang DS, Jonker JW, Kato Y, et al.: Involvement of
                                                                     organic cation transporter 1 in hepatic and intestinal
data may produce an unexpected change in direction.                  distribution of metformin. J Pharmacol Exp Ther 2002,
Watch this space!                                                    302:510–515.
180
      I   Genetics

12.     Kimura N, Masuda S, Tanihara Y, et al.: Metformin is a             29.     Gloyn AL, Hashim Y, Ashcroft SJ, et al.: Association studies
        superior substrate for renal organic cation transporter OCT2               of variants in promoter and coding regions of beta-cell
        rather than hepatic OCT1. Drug Metab Pharmacokinet                         ATP-sensitive K-channel genes SUR1 and Kir6.2 with type 2
        2005, 20:379–386.                                                          diabetes mellitus (UKPDS 53). Diabet Med 2001, 18:206–212.
13.     Wang DS, Kusuhara H, Kato Y, et al.: Involvement of                30.• Grant SF, Thorleifsson G, Reynisdottir I, et al.: Variant of
        organic cation transporter 1 in the lactic acidosis caused by              transcription factor 7-like 2 (TCF7L2) gene confers risk of
        metformin. Mol Pharmacol 2003, 63:844–848.                                 type 2 diabetes. Nat Genet 2006, 38:320–323.
14.•• Shu Y, Sheardown SA, Brown C, et al.: Effect of genetic              This is a landmark study detailing the discovery of what remains
        variation in the organic cation transporter 1 (OCT1) on            the largest common type 2 diabetes risk variant to date.
        metformin action. J Clin Invest 2007, 117:1422–1431.               31.     Saxena R, Gianniny L, Burtt NP, et al.: Common single
This is an elegant paper that describes how Oct1 is necessary for                  nucleotide polymorphisms in TCF7L2 are reproducibly
metformin to elicit a therapeutic effect in mice lacking Oct1; loss-               associated with type 2 diabetes and reduce the insulin
of-function human polymorphisms are then described that reduce                     response to glucose in nondiabetic individuals. Diabetes
metformin action in 20 glucose-tolerant humans.                                    2006, 55:2890–2895.
15.     Shu Y, Brown C, Castro RA, et al.: Effect of genetic variation     32.     Lyssenko V, Lupi R, Marchetti P, et al.: Mechanisms by
        in the organic cation transporter 1, OCT1, on metformin                    which common variants in the TCF7L2 gene increase risk
        pharmacokinetics. Clin Pharmacol Ther 2008, 83:273–280.                    of type 2 diabetes. J Clin Invest 2007, 117:2155–2163.
16.     Kirchheiner J, Thomas S, Bauer S, et al.: Pharmacokinetics and     33.     Florez JC, Jablonski KA, Bayley N, et al.: TCF7L2
        pharmacodynamics of rosiglitazone in relation to CYP2C8                    polymorphisms and progression to diabetes in the Diabetes
        genotype. Clin Pharmacol Ther 2006, 80:657–667.                            Prevention Program. N Engl J Med 2006, 355:241–250.
17.     Tornio A, Niemi M, Neuvonen PJ, Backman JT: Trim-                  34.• Pearson ER, Donnelly LA, Kimber C, et al.: Variation in
        ethoprim and the CYP2C8*3 allele have opposite effects on                  TCF7L2 influences therapeutic response to sulfonylureas:
        the pharmacokinetics of pioglitazone. Drug Metab Disp                      a GoDARTs study. Diabetes 2007, 56:2178–2182.
        2008, 36:73–80.                                                    This is a large observational study showing that TCF7L2 variants
18.     Pearson ER, Starkey BJ, Powell RJ, et al.: Genetic cause           affect sulfonylurea but not metformin response.
        of hyperglycaemia and response to treatment in diabetes.           35.• Natali A, Ferrannini E: Effects of metformin and thiazoli-
        Lancet 2003, 362:1275–1281.                                                dinediones on suppression of hepatic glucose production
19.     Shepherd M, Pearson ER, Houghton J, et al.: No deterioration               and stimulation of glucose uptake in type 2 diabetes: a
        in glycemic control in HNF-1alpha maturity-onset diabetes                  systematic review. Diabetologia 2006, 49:434–441.
        of the young following transfer from long-term insulin to          This is a comprehensive review of the physiologic data on metformin
        sulphonylureas. Diabetes Care 2003, 26:3191–3192.                  and thiazolidinedione action.
20.     Gloyn AL, Pearson ER, Antcliff JF, et al.: Activating              36.     Horikoshi M, Hara K, Ohashi J, et al.: A polymorphism in
        mutations in the gene encoding the ATP-sensitive potassium-                the AMPKalpha2 subunit gene is associated with insulin
        channel subunit Kir6.2 and permanent neonatal diabetes.                    resistance and type 2 diabetes in the Japanese population.
        N Engl J Med 2004, 350:1838–1849.                                          Diabetes 2006, 55:919–923.
21.     Proks P, Arnold AL, Bruining J, et al.: A heterozygous             37.     Sun MW, Lee JY, de Bakker PI, et al.: Haplotype structures
        activating mutation in the sulfphonylurea receptor SUR1                    and large-scale association testing of the 5’ AMP-activated
        (ABCC8) causes neonatal diabetes. Hum Mol Genet 2006,                      protein kinase genes PRKAA2, PRKAB1, and PRKAB2
        15:1793–1800.                                                              [corrected] with type 2 diabetes. Diabetes 2006, 55:849–855.
22.     Babenko AP, Polak M, Cave H, et al.: Activating mutations in       38.     Keshavarz P, Inoue H, Nakamura N, et al.: Single nucleotide
        the ABCC8 gene in neonatal diabetes mellitus. N Engl J Med                 polymorphisms in genes encoding LKB1 (STK11), TORC2
        2006, 355:456–466.                                                         (CRTC2) and AMPK alpha2-subunit (PRKAA2) and risk of
23.     Sagen JV, Raeder H, Hathout E, et al.: Permanent neonatal                  type 2 diabetes. Mol Genet Metab 2008, 93:200–209.
        diabetes due to mutations in KCNJ11 encoding Kir6.2:               39.     Legro RS, Barnhart HX, Schlaff WD, et al.: Ovulatory
        patient characteristics and initial response to sulfonylurea               response to treatment of polycystic ovary syndrome is
        therapy. Diabetes 2004, 53:2713–2718.                                      associated with a polymorphism in the STK11 gene. J Clin
24.     Pearson ER, Flechtner I, Njolstad PR, et al.: Switching from               Endocrinol Metab 2008, 93:792–800.
        insulin to oral sulfonylureas in patients with diabetes due to     40.     Yki-Jarvinen H: Thiazolidinediones. N Engl J Med 2004,
        Kir6.2 mutations. N Engl J Med 2006, 355:467–477.                          351:1106–1118.
25.     Frayling TM: Genome-wide association studies provide               41.     Altshuler D, Hirschhorn JN, Klannemark M, et al.: The
        new insights into type 2 diabetes etiology. Nat Rev 2007,                  common PPARgamma Pro12Ala polymorphism is associated
        8:657–662.                                                                 with decreased risk of type 2 diabetes. Nat Genet 2000,
26.•• Feng Y, Mao G, Ren X, et al.: Ser1369Ala variant in                          26:76–80.
        sulfonylurea receptor gene ABCC8 is associated with                42.     Muller YL, Bogardus C, Beamer BA, et al.: A functional
        antidiabetic efficacy of gliclazide in Chinese type 2 diabetic             variant in the peroxisome proliferator-activated receptor
        patients. Diabetes Care 2008, 31:1939–1944.                                gamma2 promoter is associated with predictors of obesity
This is probably the largest prospective study of genetics of                      and type 2 diabetes in Pima Indians. Diabetes 2003,
sulfonylurea response, describing (with replication) a variant in                  52:1864–1871.
ABCC8 (encoding the sulfonylurea receptor SUR1) that influences            43.     Bluher M, Lubben G, Paschke R: Analysis of the relationship
gliclazide response.                                                               between the Pro12Ala variant in the PPAR-gamma2 gene
27.     Gloyn AL, Weedon MN, Owen KR, et al.: Large-scale                          and the response rate to therapy with pioglitazone in patients
        association studies of variants in genes encoding the pancreatic           with type 2 diabetes. Diabetes Care 2003, 26:825–831.
        beta-cell KATP channel subunits Kir6.2 (KCNJ11) and SUR1           44.     Snitker S, Watanabe RM, Ani I, et al.: Changes in
        (ABCC8) confirm that the KCNJ11 E23K variant is associated                 insulin sensitivity in response to troglitazone do not
        with type 2 diabetes. Diabetes 2003, 52:568–572.                           differ between subjects with and without the common,
28.     Sesti G, Laratta E, Cardellini M, et al.: The E23K variant                 functional Pro12Ala peroxisome proliferator-activated
        of KCNJ11 encoding the pancreatic beta-cell adenosine                      receptor-gamma2 gene variant: results from the Tro-
        5’-triphosphate-sensitive potassium channel subunit Kir6.2                 glitazone in Prevention of Diabetes (TRIPOD) study.
        is associated with an increased risk of secondary failure                  Diabetes Care 2004, 27:1365–1368.
        to sulfonylurea in patients with type 2 diabetes. J Clin
        Endocrinol Metab 2006, 91:2334–2339.
Pharmacogenetics in Diabetes
                                                                                                                         I   Pearson
                                                                                                                                       I   181

45.   Wolford JK, Yeatts KA, Dhanjal SK, et al.: Sequence              53.     Hansen L, Ekstrom CT, Tabanera YPR, et al.: The
      variation in PPARG may underlie differential response to                 Pro12Ala variant of the PPARG gene is a risk factor for
      troglitazone. Diabetes 2005, 54:3319–3325.                               peroxisome proliferator-activated receptor-gamma/alpha
46.   Florez JC, Jablonski KA, Sun MW, et al.: Effects of the                  agonist-induced edema in type 2 diabetic patients. J Clin
      type 2 diabetes-associated PPARG P12A polymorphism on                    Endocrinol Metab 2006, 91:3446–3450.
      progression to diabetes and response to troglitazone. J Clin     54.•• Link E, Parish S, Armitage J, et al.: SLCO1B1 variants and
      Endocrinol Metab 2007, 92:1502–1509.                                     statin-induced myopathy—a genomewide study. N Engl J Med
47.   Kang ES, Park SY, Kim HJ, et al.: Effects of Pro12Ala poly-              2008, 359:789–799.
      morphism of peroxisome proliferator-activated receptor           This paper discussed the fi rst published genome-wide scan applied
      gamma2 gene on rosiglitazone response in type 2 diabetes.        to drug response showing that it is possible to detect large adverse
      Clin Pharmacol Ther 2005, 78:202–208.                            effects with a small sample size. Variants in SLCO1B1 are associ-
48.   Kang ES, Park SY, Kim HJ, et al.: The influence of adiponectin   ated with severe myopathy in users of high-dose statins.
      gene polymorphism on the rosiglitazone response in patients      55.     Holstein A, Plaschke A, Ptak M, et al.: Association between
      with type 2 diabetes. Diabetes Care 2005, 28:1139–1144.                  CYP2C9 slow metabolizer genotypes and severe hypogly-
49.   Sun H, Gong ZC, Yin JY, et al.: The association of                       caemia on medication with sulphonylurea hypoglycaemic
      adiponectin allele 45T/G and -11377C/G polymorphisms                     agents. Br J Clin Pharmacol 2005, 60:103–106.
      with Type 2 diabetes and rosiglitazone response in Chinese       56.     Sesti G, Marini MA, Cardellini M, et al.: The Arg972
      patients. Br J Clin Pharmacol 2008, 65:917–926.                          variant in insulin receptor substrate-1 is associated with
50.   Brunham LR, Kruit JK, Pape TD, et al.: Beta-cell ABCA1                   an increased risk of secondary failure to sulfonylurea
      influences insulin secretion, glucose homeostasis and                    in patients with type 2 diabetes. Diabetes Care 2004,
      response to thiazolidinedione treatment. Nat Med 2007,                   27:1394–1398.
      13:340–347.                                                      57.     Kang ES, Cha BS, Kim HJ, et al.: The 11482G >A polymor-
51.   Wang J, Bao YQ, Hu C, et al.: Effects of ABCA1 variants                  phism in the perilipin gene is associated with weight gain
      on rosiglitazone monotherapy in newly diagnosed type 2                   with rosiglitazone treatment in type 2 diabetes. Diabetes
      diabetes patients. Acta Pharmacol Sin 2008, 29:252–258.                  Care 2006, 29:1320–1324.
52.   Spraggs C, McCarthy A, McCarthy L, et al.: Genetic               58.     Wang G, Wang X, Zhang Q, Ma Z: Response to pioglitazone
      variants in the epithelial sodium channel associate with                 treatment is associated with the lipoprotein lipase S447X
      oedema in type 2 diabetic patients receiving the peroxisome              variant in subjects with type 2 diabetes mellitus. Int J Clin
      proliferator-activated receptor gamma agonist farglitazar.               Pract 2007, 61:552–557.
      Pharmacogenet Genomics 2007, 17:1065–1076.
You can also read