BON-10693; No. of pages: 8; 4C: Bone xxx (2015) xxx–xxx

Contents lists available at ScienceDirect

Bone

1

Review

2Q1

Effects of diabetes drugs on the skeleton

3Q2

Christian Meier a,⁎, Ann V. Schwartz b, Andrea Egger a, Beata Lecka-Czernik c,d

4 5 6 7

a

8

a r t i c l e

9 10 11 12 13

Article history: Received 19 March 2015 Revised 13 April 2015 Accepted 16 April 2015 Available online xxxx

14 15 16 17 18 19 20

Keywords: Diabetes mellitus Metformin TZDs Incretins DPP-4 inhibitors SGLT2 inhibitors

F

journal homepage: www.elsevier.com/locate/bone

Division of Endocrinology, Diabetes and Metabolism, University Hospital, Basel, Switzerland Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA c Department of Orthopedic Surgery, Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, OH, USA d Department of Physiology and Pharmacology, Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, OH, USA

i n f o

R O

O

b

a b s t r a c t

R

E

C

T

E

D

P

Type 2 diabetes is associated with increased fracture risk and the mechanisms underlying the detrimental effects of diabetes on skeletal health are only partially understood. Antidiabetic drugs are indispensable for glycemic control in most type 2 diabetics, however, they may, at least in part, modulate fracture risk in exposed patients. Preclinical and clinical data clearly demonstrate an unfavorable effect of thiazolidinediones on the skeleton with impaired osteoblast function and activated osteoclastogenesis. The negative effect of thiazolidinediones on osteoblastogenesis includes decreased activity of osteoblast-specific transcription factors (e.g. Runx2, Dlx5, osterix) and decreased activity of osteoblast-specific signaling pathways (e.g. Wnt, TGF-β/BMP, IGF-1). In contrast, metformin has a positive effect on osteoblast differentiation due to increased activity of Runx2 via the AMPK/USF-1/ SHP regulatory cascade resulting in a neutral or potentially protective effect on bone. Recently marketed antidiabetic drugs include incretin-based therapies (GLP-1 receptor agonists, DPP-4 inhibitors) and sodium-glucose co-transporter 2 (SGLT2)-inhibitors. Preclinical studies indicate that incretins (GIP, GLP-1, and GLP-2) play an important role in the regulation of bone turnover. Clinical safety data are limited, however, meta-analyses of trials investigating the glycemic-lowering effect of both, GLP-1 receptor agonists and DPP4-inhibitors, suggest a neutral effect of incretin-based therapies on fracture risk. For SGLT2-inhibitors recent data indicate that due to their mode of action they may alter calcium and phosphate homeostasis (secondary hyperparathyroidism induced by increased phosphate reabsorption) and thereby potentially affect bone mass and fracture risk. Clinical studies are needed to elucidate the effect of SGLT2-inhibitors on bone metabolism. Meanwhile SGLT2-inhibitors should be used with caution in patients with high fracture risk, which is specifically true for the use of thiazolidinediones. © 2015 Published by Elsevier Inc.

R

40 41 45 43 42

48 49 50 51 52 53 54 55 56 57 58

Contents

Introduction . . . . . . . . . . . . . . . . Metformin . . . . . . . . . . . . . . . . . Thiazolidinediones . . . . . . . . . . . . . Mechanism of TZD-induced bone loss . . Novel selective PPARγ modulators with Sulfonylureas . . . . . . . . . . . . . . . . Incretin-based therapies . . . . . . . . . . . GLP-1 receptor agonist . . . . . . . . . DPP-4 inhibitors . . . . . . . . . . . . SGLT-2 inhibitors . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . .

U

47 46

N C O

44

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . beneficial effect of . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . insulin sensitizers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . and no . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . effect . . . . . . . . . . . . . . . . . .

. . . . . . . . on . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . adipocyte . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . differentiation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

. . . . . . . . . . .

. . . . . . . . . . .

. . . . . . . . . . .

. . . . . . . . . . .

. . . . . . . . . . .

. . . . . . . . . . .

. . . . . . . . . . .

. . . . . . . . . . .

59

⁎ Corresponding author at: Division of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Missionsstrasse 24, CH-4055 Basel, Switzerland. Fax: +41 61 264 97 96. E-mail address: [email protected] (C. Meier).

http://dx.doi.org/10.1016/j.bone.2015.04.026 8756-3282/© 2015 Published by Elsevier Inc.

Please cite this article as: Meier C, et al, Effects of diabetes drugs on the skeleton, Bone (2015), http://dx.doi.org/10.1016/j.bone.2015.04.026

0 0 0 0 0 0 0 0 0 0 0

21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39

C. Meier et al. / Bone xxx (2015) xxx–xxx

82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107 108 109 110

C

80 81

E

78 79

R

76 77

R

74 75

O

72 73

C

70 71

N

68 69

U

66 67

Metformin

124

F

Table 1 Antidiabetic drugs and their effect on fracture risk.

65

Metformin is most commonly used to increase insulin sensitivity in diabetic patients. Biguanides decrease hepatic glucose production and increase glucose uptake in muscle. Metformin is considered by the World Health Organization an essential medicine satisfying the criteria of the public health relevance, evidence on efficacy and safety, and comparative cost effectiveness (www.who.int/medicines). Metformin mechanism of insulin sensitization includes activation of hepatic and muscle AMP-activated protein kinase (AMPK), which results in suppression of fatty acid synthesis and stimulation of fatty acid oxidation in liver and increase in muscle glucose uptake [26]. AMPK also decreases expression of sterol-regulatory element-binding-protein 1 (SREBP-1), a transcription factor involved in adipocyte differentiation and pathogenesis of insulin resistance, dyslipidemia and diabetes. Animal studies indicate that metformin has a positive effect on osteoblast differentiation due to increased activity of osteoblast-specific Runx2 transcription factor via AMPK/USF-1/SHP regulatory cascade [27] and it has a negative effect on osteoclast differentiation and bone loss after ovariectomy by decreasing RANKL and increasing osteoprotegerin levels [28]. Interestingly, in rodent models metformin can prevent the adverse effects of TZDs on bone by either inducing re-ossification of bone after rosiglitazone treatment or preventing rosiglitazone effects when applied in combination with rosiglitazone [29]. There are few clinical studies investigating the effect of metformin on bone and fracture risk. Metformin was compared with a sulfonylurea (glyburide) and with a thiazolidinedione (rosiglitazone) in the ADOPT trial, discussed in more detail in the section on thiazolidinediones [30]. The primary endpoint of this trial was time to monotherapy failure. Fractures were identified as adverse events. Fracture incidence was similar in those assigned to metformin or glyburide. One-year changes in the bone resorption marker CTX were similar in women (difference in 12-month change: + 2.0%) and modestly greater in men (− 8.4%) in those assigned to metformin compared with a sulfonylurea [31]. The metformin group had greater decreases in levels of the bone formation marker P1NP (difference in 12-month change: −9.4% women; −19.5% men), compared with the sulfonylurea group. Most observational studies on metformin have found no effect on fracture risk although three studies have reported reduced risk [32–34]. Metformin use was associated with lower fracture risk,

O

t1:1 t1:2

63 64

R O

111

Type 2 diabetes is associated with increased fracture risk despite the fact that patients with diabetes have higher bone mineral density as compared to non-diabetic individuals [1–3]. The mechanisms underlying the detrimental effects of diabetes on skeletal health are only partially understood. It is assumed that determinants of fracture risk are multifactorial including diabetes-related microvascular complications, fall risk and alterations associated with chronic hyperglycemia [4]. As documented in preclinical models hyperglycemia may alter calcium and vitamin D metabolism resulting in impaired bone mineralization [5,6]. Furthermore, chronic hyperglycemia may result in deposition of advanced glycosylation end-products in bone collagen (such as pentosidine) contributing to impaired bone quality [7,8] and higher fracture risk [9,10]. Several studies suggest that skeletal dynamics are reduced in type 2 diabetes [4] with decreased osteoblast function as documented by reduced biochemical markers of bone formation [11] and lower bone formation rate in a histomorphometric study [12]. Several pathophysiological changes in diabetics might contribute to decreased bone formation. They include interference of advanced glycosylation end-products with osteoblast development [13], function [14] and attachment to collagen matrix [15], increased levels of osteocytederived sclerostin [16–18], and hyperglycemia-induced suppression of osteogenic differentiation of marrow-derived progenitor cells diverting osteoblastic precursor cells to a metabolically stressed adipogenic pathway that induces synthesis of a hyaluronan matrix that recruits inflammatory cells and establishes an inflammatory process contributing to bone demineralization [19]. Antidiabetic drugs are indispensable for glycemic control in most type 2 diabetics. However before discussing potential benefits or risks of antidiabetic drugs on bone metabolism it seems evident that optimal glycemic control per se is an important contributing factor for improvement of skeletal integrity in diabetic patients. This notion is supported by several studies showing increased fracture risk in patients with poor glycemic control and reduced risk in patients on intensive glycemic control. A recent cohort study explored the association between glycemic control as measured by serum hemoglobin A1c (HbA1c) levels and the risk of hip fracture in type 2 diabetics aged over 65 years and observed a linear relationship between HbA1c and hip fracture risk. After adjustment for various contributing factors hip fracture risk was 24–31% higher among diabetics with HbA1c levels above 9% than among patients with HbA1c levels of 6–7% [20]. These data are in line with some but not all previous studies confirming a detrimental effect of poor glycemic control on fracture risk [21–23]. In contrast, however, this relationship could not be observed in the ACCORD trial, a clinical trial investigating type 2 diabetics randomized either to intensive or standard treatment strategies. The lack of significant effect of glycemic control on the occurrence of non-vertebral fractures (and falls) might be attributed to the small difference in effective diabetes control between patients with intensified treatment strategy (HbA1c 6.4%) and standard treatment (HbA1c 7.5%) [24]. Although reducing hyperglycemia is mandatory not only for skeletal health but also in decreasing

112 113

P

61 62

the onset and progression of microvascular complications, individualized treatment is necessary, balancing the benefits and risks of glycemic control based on the patient's age and health status [25]. Drug-induced hypoglycemic episodes need to be avoided which in addition to diabetic complications (neuropathy, retinopathy) may increase the risk of falls and fractures. This review summarizes the effects of antidiabetic drugs on bone metabolism and fracture risk (Table 1). Preclinical and clinical data of both, insulin sensitizers (metformin, thiazolidinediones) and insulin secretagogues are discussed with specific focus on the skeletal effects of recently marketed drugs such as incretin-based therapies (GLP-1 receptor agonists, DPP-4 inhibitors) and SGLT2-inhibitors.

D

Introduction

T

60

E

2

t1:3

Target

Mode of action

Class of drugs

Drugs

Fracture Risk

t1:4 t1:5 t1:6 t1:7 t1:8 t1:9 t1:10 t1:11 t1:12

Insulin

Sensitizers

Biguanides Thiazolidinediones K + ATP GLP-1 analogs

Metformin Pioglitazone, rosiglitazone Sulfonylureas Liraglutide Exenatide Sitagliptin, etc. Acarbose, miglitol, voglibose Pramlintide Canagliflozin, dapagliflozin

↓ ↑ Neutral Neutral - ↓ Neutral - ↑ Neutral - ↓ ? ? (↑)

Secretagogues

DPP-4 inhibitors Other

α-glucosidase inhibitors Amylin analog SGLT2 inhibitors

Please cite this article as: Meier C, et al, Effects of diabetes drugs on the skeleton, Bone (2015), http://dx.doi.org/10.1016/j.bone.2015.04.026

114 115 116 117 118 119 120 121 122 123

125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162

C. Meier et al. / Bone xxx (2015) xxx–xxx

187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222 223 224 225 226

C

185 186

E

183 184

R

181 182

R

179 180

N C O

177 178

U

Mechanism of TZD-induced bone loss

275

PPARγ, an essential regulator of lipid, glucose, and insulin metabolism [62], is a target for TZDs. The PPARγ protein is expressed in mice and humans in two isoforms, PPARγ1 and PPARγ2. PPARγ1 is expressed in a variety of cell types, including cells of mesenchymal and hematopoietic lineage, whereas PPARγ2 expression is restricted to cells of mesenchymal lineage adipocytes. In bone, PPARγ2 plays an important role in regulation of mesenchymal stem cell (MSC) differentiation toward osteoblasts and adipocytes, and the maintenance of bone mass. In marrow MSCs, PPARγ2 activated with rosiglitazone induces adipocyte differentiation, suppresses osteoblast differentiation, and increases RANKL expression and support for osteoclastogenesis [63–65]. In contrast, PPARγ1 expressed in hematopoietic stem cells promotes osteoclast differentiation and bone resorption [66]. It controls an expression of c-fos protein, an important determinant of osteoclast lineage commitment and development, and activates Pgc1β, an important co-

276 277

F

175 176

O

TZDs increase insulin sensitivity via activation of peroxisome proliferator-activated receptor (PPARγ). Two TZDs, rosiglitazone and pioglitazone, have been used clinically since 1999. A number of studies showed superior efficacy of TZDs over other available antidiabetic therapies in the control of diabetic hyperglycemia [41]. However, their prolonged use is associated with several adverse effects. Strong clinical evidence points to the connection between rosiglitazone use and a significant increase in risk of myocardial infarction and death from cardiovascular causes [42]. This association resulted in a recent review of rosiglitazone safety by the FDA and recommendation for its restricted use in the US. Interestingly, pioglitazone use is associated with a significantly lower risk of death and lower number of myocardial infarction and stroke incidence [43], indicating that cardiovascular effects of TZDs are not a drug class effect, but rather specifically associated with the TZD type. However, increased risk of bladder cancer in long-term pioglitazone users resulted in recent restriction of its use by FDA. Both TZDs exhibit drug class properties of fluid retention and weight gain [44]. Although the use of both rosiglitazone and pioglitazone is currently restricted, new TZDs with better safety profile are in development. Therefore, understanding TZDs mechanism of action on bone is needed in respect to improvement of safety for bone of new line of TZDs. Clinical studies of rosiglitazone and pioglitazone use have identified adverse effects on the skeleton [45,46]. The first demonstration of increased fracture risk was reported from ADOPT (A Diabetes Outcome Progression Trial), designed to compare time to monotherapy failure of rosiglitazone, metformin and glyburide in recently diagnosed T2DM patients [41]. The trial included 2511 men and 1840 women with an average age of 56 (SD 10) years. In a post hoc analysis of fracture rates, using adverse event reports to identify fractures, fracture incidence in men did not differ across treatment groups [30,41]. However, in women, fracture incidence with rosiglitazone was approximately doubled compared with the other two treatments. The hazard ratios for fractures in women were 1.81 (95% CI 1.17–2.80) and 2.13 (95% CI 1.30–3.51) for rosiglitazone compared with metformin and glyburide, respectively. Increased fracture rates were seen in the lower and upper limbs. Hip and vertebral fractures did not differ across treatment assignments, but only 4 hip and 3 vertebral fractures were reported in women. The other prescribed TZD, pioglitazone, is also associated with increased fracture risk in women, but not men. This finding was first reported by the manufacturer based on a meta-analysis of pioglitazone trials [47]. Two subsequent meta-analyses of randomized trials have supported these initial findings of increased fracture risk with rosiglitazone and pioglitazone use in women without evidence of elevated risk in men [48,49]. The more recent meta-analysis, based on 22 RCTs, reported that effects in women were similar for rosiglitazone (OR = 2.10; 95% CI 1.61–2.51) and pioglitazone (OR = 1.73; 95% CI 1.18–2.55) [49]. The TZD trials included few participants over 70 years old, the age range when hip fractures become more common. The numbers of hip fractures in the trials are not sufficient to assess them separately. Observational studies have reported increased risk of hip fracture with TZD use. A recent study using registry data in Scotland found increased hip

170 171

R O

174

169

227 228

P

Thiazolidinediones

167 168

fracture with greater cumulative TZD use among those with any use (OR per year of exposure 1.18; 95% CI 1.09–1.28) [38]. Results were similar for pioglitazone and rosiglitazone considered separately. In contrast to reports from randomized trials, increased hip fracture risk was found in men (OR per year of exposure 1.20; 95% CI 1.03–1.41) as well as in women (OR per year of exposure 1.18; 95% CI 1.07–1.29). Two large observational studies using data from the UK also reported that TZD use increased hip fracture [34,50]. Most [45,51–56], but not all [57,58], randomized trials testing the effects of TZD use on bone density have found more rapid bone loss under therapy. A recent meta-analysis of ten randomized clinical trials that assessed change in BMD reported greater bone loss at the lumbar spine, total hip and femoral neck in women randomized to TZD treatment compared with placebo or other antidiabetic medication [49]. Assessments of changes in bone turnover markers with TZD treatment in clinical trials have not been entirely consistent but suggest modest increase in resorption and possibly decrease in formation. In the largest study to date, 12-month changes in serum markers were assessed in 1605 participants in the ADOPT trial [31]. Among women, rosiglitazone use was associated with increases in the resorption marker C-terminal telopeptide (CTX) compared with glyburide (10.7% difference, p = 0.002) or metformin (7.3% difference, p = 0.029). In men, CTX was elevated in the rosiglitazone group compared with metformin (12.2%, p b 0.001) but not compared with glyburide. Women and men had modest reductions in the bone formation marker P1NP (women − 4.4%, men − 14.4%), but those in the metformin arm experienced greater reductions. For women, changes in P1NP did not differ between the rosiglitazone and glyburide groups while in men losses were greater in the rosiglitazone group. Smaller trials of rosiglitazone treatment have also reported relative increases in markers of bone resorption compared to placebo [57,59] and to metformin [55]. These results indicate that increases in bone resorption may explain at least in part the increased fracture rate in women on TZD therapy [31]. Some smaller trials have reported a relative reduction in bone formation markers with rosiglitazone treatment, compared with placebo [60] or with diet only treatment [53], and others have reported no difference [54]. For pioglitazone, the largest trial included 156 postmenopausal women with pre-diabetes and found no differences in bone turnover markers after 12 months compared with placebo [58]. In contrast, a trial in 71 diabetic men reported relative increases in markers of bone resorption (CTX) and formation (P1NP) in the pioglitazone group compared with metformin [61] while a trial in 86 diabetic men and women found a relative increase in a formation but not a resorption marker with pioglitazone treatment compared with placebo [56]. In summary, clinical studies have found that TZD use is associated with increased fracture risk in women and possibly in men. This increased risk appears to be a class effect of both rosiglitazone and pioglitazone. More rapid bone loss seems to be the underlying mechanism.

D

173

165 166

T

172

compared with non-diabetic residents, in a Danish population [33]. In the Rochester MN population metformin use was associated with a lower rate of fracture compared with T2DM patients who were not using metformin (adjusted hazard ratio 0.7; 95% CI 0.6–0.96) [32]. On the other hand, several studies have reported no difference in fracture risk with metformin use [35–40]. A recent report on diabetes and hip fracture found that metformin tends to be prescribed to patients with a lower overall risk of fracture [38]. This prescribing pattern may account for some of the reduced fracture risk associated with metformin use in observational studies.

E

163 164

3

Please cite this article as: Meier C, et al, Effects of diabetes drugs on the skeleton, Bone (2015), http://dx.doi.org/10.1016/j.bone.2015.04.026

229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269 270 271 272 273 274

278 279 280 281 282 283 284 285 286 287 288 289 290

316 317 318 319 320 321 322 323 324 325 326 327

332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353

C

314 315

E

312 313

R

310 311

R

308 309

O

306 307

C

304 305

N

302 303

U

300 301

F

The PPARγ ligand-binding domain contains a large binding pocket capable of encompassing a variety of ligands. This provides a wide array of potential contact points that can result in various PPARγ post-translational modifications (PTMs), including phosphorylation, acetylation and sumoylation, and differential recruitment of coactivators, which determine specific activities of this nuclear receptor [78]. The molecular studies provide evidence for distinct mechanisms regulating the proadipocytic, antiosteoblastic, and insulin sensitizing activities of PPARγ and include the levels of Serine 273 and Serine 112 phosphorylation and functional interaction with other proteins such as β-catenin and molecular chaperons FKBP51 and PP5 [79–82]. The concern of TZDs' adverse effects has prompted pharmaceutical efforts to develop selective PPARγ modulators which will retain high potency to treat diabetic disease with minimal adverse effects [83]. The PPARγ selective activators, with a decreased proadipocytic activity but intact insulin sensitizing activity such as netoglitazone, INT131, MSDC-0602 and telmisartan do not affect bone mass in mice treated with the therapeutic doses [84–87]. A new class of insulin sensitizers with structural similarities to telmisartan, which block Serine 273 phosphorylation but do not stimulate PPARγ transcriptional proadipocytic activity, has been recently developed [79,88], however their safety for bone is not as yet determined.

298 299

355 356

O

331

297

Although sulfonylureas are widely used for many years in type 2 diabetics, data on their skeletal safety are scarce. Nevertheless, based on epidemiological studies with adjustments for covariables, sulfonylureas seem to have a beneficial effect on fracture risk irrespective of the duration of treatment [34,89]. Evidence from both the ADOPT studies and the Rochester studies indicates that glibenclamide (glyburide) therapy does not have an effect on bone mass and fracture risk [32,41]. Within the ADOPT trial the cumulative incidence of fractures was comparable in glibenclamide and metformin users and significantly lower as compared to rosiglitazone users [30]; however, glibenclamide treatment decreased serum levels of bone formation markers (P1NP) in the ADOPT study [90,91]. Hypoglycemia is a frequent complication of sulfonylurea use, specifically in elderly patients treated with glibenclamide. In the UK Prospective Diabetes Study up to 18% of type 2 diabetics on sulfonylureas experienced hypoglycemia [92]. As hypoglycemia may lead to falls, occurrence of fall-related, osteoporotic fractures in elderly is assumed. A systematic review with the purpose to review the literature regarding the use of sulfonylurea and falls or fall-related fractures among older type 2 diabetics was recently published [93]. Lapane et al. conclude that data available from existing studies suffer methodological limitations, because these studies were not designed to evaluate the risk of sulfonylureas on fractures and falls in elderly adults. Hence, the risk of falls and fractures may be underestimated in users of sulfonylureas [93].

R O

Novel selective PPARγ modulators with beneficial effect of insulin sensitizers and no effect on adipocyte differentiation

295 296

354

P

329 330

293 294

Sulfonylureas

Incretin-based therapies Nearly 10 years ago glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors have been approved for the treatment of type 2 diabetes and are widely used as second-line agents in case of inadequate glycemic control [25]. Incretins are gut-derived hormones which exert their actions through activation of incretin receptor signaling. Glucagon-like peptides (GLP-1, GLP-2) and glucose-dependent insulinotropic polypeptide (GIP) are related hormones released from intestinal cells in response to nutrient intake. Although GLP-1 and GLP-2 are synthesized and secreted together from enteroendocrine cells they exhibit different biological functions. GLP-1 receptors are highly expressed on islet β cells, but also widely in non-islet cells promoting metabolic effect in various organs including bone [94]. In contrast, the GLP-2 receptors are expressed within the gastrointestinal tract. The predominant role for GLP-2 is its promotion of growth and function of intestinal mucosa, including gut barrier function and cytoprotective functions in the small bowel [95,96]. Bioactivity of incretin hormones is limited by their rapid degradation and inactivation by dipeptidyl peptidase-4, a serine protease that is present in a soluble form in plasma and is expressed in most tissues [97]. This has led to the development of degradationresistant GLP-1 receptor agonists and DPP-4 inhibitors. As GLP-1 receptors are found in various tissues, their functional effects are multifactorial. Favorable metabolic effects of GLP-1 include enhancement of glucose-stimulated insulin secretion, inhibition of glucose-dependent glucagon secretion and control of appetite and body weight [98,99]. Bone cells, including osteoblasts and osteoclasts have been shown to express receptors for both GIP and GLP incretins. A number of studies indicate that GLP-2 acts mainly as an anti-resorptive [100], whereas GIP can act as an anti-resorptive and anabolic hormone [101,102]. Furthermore, GLP-1 receptor has been shown to be essential for control of bone resorption as mice deficient in GLP-1 receptor present with cortical porosity as a result of increased osteoclastic bone resorption activity. In contrast to direct inhibitory effects of GIP and GLP-2 on bone resorption, the effect of GLP-1 is indirect through a calcitonindependent pathway [103].

T

328

factor for energy utilization by the osteoclast [67]. PPARγ1 is also expressed in osteoblasts and negatively regulates their differentiation through suppression of mTOR signaling [68]. An essential role of PPARγ in maintenance of bone homeostasis was demonstrated in several animal models of either bone accrual or bone loss depending on the status of PPARγ activity [69–74]. In models of bone accrual, a decrease in PPARγ activity in either heterozygous PPARγ-deficient mice or mice carrying a hypomorphic mutation in the PPARγ gene locus or mice with targeted PPARγ deletion in either mesenchymal cells or osteoblasts led to increased bone mass due to increased quantity of osteoblasts [68,72,74,75]. Interestingly, mice deficient in PPARγ expression in cells of hematopoietic lineage develop osteopetrosis and are less sensitive to the TZD-induced bone loss than control mice [66]. In contrast, PPARγ activation with full agonist rosiglitazone resulted in significant decreases in BMD, bone volume and changes in bone microarchitecture [65,69–71,73]. Observed bone loss was associated with changes in the structure and function of bone marrow, which included decreased number of osteoblasts, and increased number of osteoclasts and adipocytes. The degree of bone loss in response to rosiglitazone correlated with the animal age and the level of PPARγ expression. In younger animals with less PPARγ, bone loss was less extensive than in older animals [65]. Moreover, age determined the mechanism by which bone loss occurred. In younger animals it occurred due to decreased bone formation, whereas in older animals due to increased bone resorption [65]. Interestingly, estrogen deficiency predisposes to the rosiglitazone-induced bone loss due to increased bone resorption [70]. In conclusion, animal studies suggest that aging and estrogen deficiency confound TZD-induced bone loss and determine its mechanism. The negative effect of TZDs on osteoblastogenesis includes decreased activity of Runx2, Dlx5, and Osterix, which are osteoblastspecific transcription factors, and decreased activity of osteoblastspecific signaling pathways controlling bone homeostasis, among them Wnt, TGF-β/BMP and IGF-1 [64,76]. The effect of TZDs on the expression of genes essential for osteoblast development is strikingly similar to changes observed during aging. Due to the type of bone loss and similarities to aging, some speculate that TZDs may accelerate the aging of bone [65,77].

D

291 292

C. Meier et al. / Bone xxx (2015) xxx–xxx

E

4

Please cite this article as: Meier C, et al, Effects of diabetes drugs on the skeleton, Bone (2015), http://dx.doi.org/10.1016/j.bone.2015.04.026

357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 375 376 377 378

379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415

C. Meier et al. / Bone xxx (2015) xxx–xxx

437 438 439 440 441 442 443 444

449 450 451 452 453 454 455 Q3 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479

C

435 436

E

433 434

R

F

431 432

R

429 430

N C O

427 428

U

425 426

O

Exendin-4 is a peptide analogue of GLP-1 and has similar properties to GLP-1. It is resistant to degradation by DPP-4 and is characterized by a high in vivo potency and a prolonged duration of action due to its longer plasma half-life compared to GLP-1 [107]. While exenatide shares about 53% of its amino acid sequence with human naive GLP-1, the long-acting human GLP-1 analog liraglutide is characterized by a much higher homology with human naive GLP-1 (97% homogeneity) (33, 34). Ma et al. evaluated the effects of exendin-4 on ovariectomy-induced osteoporosis in old rats. Sixteen weeks of treatment with exendin-4 enhanced bone strength and prevented the deterioration of trabecular microarchitecture. Gene expression results showed that exendin-4 not only inhibited bone resorption by increasing OPG/RANKL ratio, but also promoted bone formation by increasing the expression of osteoblast-specific transcription factors [108]. Preclinical data are documenting that GLP-1 receptor is present on MLO-Y4 cells and osteocytes of rat femurs further supporting the osteoanabolic effect of GLP-1. Exendin-4 reduced the mRNA expression and protein production of SOST/sclerostin in MLO-Y4 cells and reduced serum levels of sclerostin in type 2 diabetic OLEF rats [109]. Clinical data on the safety of incretins on human bone are limited. In contrast to the above mentioned preclinical studies in rats a 44-week treatment with exendin-4 (exenatide) in patients with type 2 diabetes did not have an effect on serum levels of bone turnover markers and did not result in a significant change in BMD although exendin-4 decreased body weight by 6% [110]. Recently, two meta-analyses investigated the association between use of GLP-1 receptor agonists and fracture incidence [111,112]. Mabilleau et al. identified seven randomized clinical trials investigating the effect of GLP-1 agonists on glycemic control in which the occurrence of a fracture was disclosed as a serious adverse event. The use of GLP-1 receptor agonists (exenatide, liraglutide) did not modify the risk of fracture in type 2 diabetics as compared to

423 424

DPP-4 inhibitors

R O

448

422

Dipeptidyl peptidase-4 (DPP-4) degrades incretin hormones to inactive metabolites. As the pharmacological effect of DPP-4 inhibitors is to prolong the action of GLP-1, their effect on bone is assumed to be similar to that of GLP-1. A recent preclinical study evaluated the effect of DPP-4 inhibitor sitagliptin on bone in normal and streptozotocin-induced diabetic rats. As shown by in vivo μCT trabecular bone loss, the decrease in trabecular number and the increase in trabecular spacing were attenuated through sitagliptin treatment in diabetic rats most likely through the reduction of bone resorption. Furthermore sitagliptin prevented cortical bone growth stagnation in diabetic rats resulting in stronger femora. All these effects were independent of glycemic control [114]. Human studies investigating the effect of DPP-4 inhibitors on bone are scarce. A neutral role of vildagliptin was demonstrated in drug naive type 2 diabetics. Postprandial circulating levels of bone resorption markers and calcium homeostasis were unaffected as compared with baseline and placebo [115]. Recently, two studies were published investigating the association between use of DPP-4 inhibitors and fracture risk [116,117]. A meta-analysis by Monami et al. of 28 clinical trials enrolling over 21,000 type 2 diabetics showed that treatment with DPP-4 inhibitors was associated with a reduced fracture risk (OR 0.60, 95% CI 0.37–0.99) compared with placebo or other antidiabetic treatments [116]. A retrospective population based cohort study using data from the Clinical Practice Research Datalink (CPRD) database, however, observed no different risk of fracture comparing current users of DPP-4 inhibitors to non-diabetic controls. In addition, fracture risk was not elevated in type 2 diabetics after adjusting for the use of other antidiabetic drugs (metformin, sulfonylurea, TZDs, and insulin) [117]. Differences between these two studies can be explained by several factors [118]. First, in the meta-analysis fractures were not routinely collected but were defined as severe side-effects in enrolled randomized trials whereas in the retrospective studies fractures were identified with high accuracy. Secondly, duration of follow-up was different with short follow-up time in the meta-analysis (mean duration, 35 weeks) and longer duration in the cohort study (median duration, 5 years). As pointed out by Driessen et al. [117], the results of their study are in keeping with a large randomized placebo-controlled study comparing cardiovascular outcomes of saxagliptin [119]. This study, not included in the meta-analysis by Monami et al. [116], showed no significant difference in the number of fractures between saxagliptin and placebo treated diabetics. In summary, incretin-based therapies seem to have an anabolic effect on bone, attenuating bone loss and potentially reducing fracture risk in type 2 diabetics. However, currently available data are not conclusive and further studies are needed to prove a potential

P

GLP-1 receptor agonist

420 421

D

447

418 419

placebo or the use of other antidiabetic medications (glimepiride, sitagliptin, and insulin) [111]. The second meta-analysis by Su et al. included 16 published or unpublished RCTs of variable duration (12 to 104 weeks) comparing the effects of exenatide or liraglutide with comparators. Separate pooled analysis was performed for exenatide and liraglutide, respectively. This meta-analysis demonstrated a divergent risk of bone fractures associated with different GLP-1 receptor agonists with exenatide increasing the risk of fractures (OR 2.09, 95% CI 1.03– 4.21) and liraglutide reducing the risk of non-vertebral fractures (OR 0.38, 95% CI 0.17–0.87), respectively (Table 1) [112]. Both studies, however, are preliminary and further studies are needed to clarify potential beneficial or detrimental effects of GLP-1 receptor agonist on fracture risk. Several methodological aspects limit the generalization of these results: both studies included randomized trials which were not designed for assessment of fracture risk and included only small numbers of fractures [113]. Furthermore, studies included in the meta-analyses were of short duration and therefore not long enough to study the treatment-effects on bone fractures.

T

445 446

Incretins play an important role in the regulation of bone turnover in response to feeding. Nutritional intake modulates bone turnover in favor of bone formation and, in contrast, fasting blunts the diurnal variation as documented by an increase of bone resorption markers. From a physiological point of view these responses are likely to have evolved to maximize skeletal strength when nutrition is abundant and to control and maintain calcium homeostasis in fasting conditions [104]. Evidence that incretins play a regulatory role in bone turnover was provided by Henriksen et al. After subcutaneous administration of GLP-2 in postmenopausal women a dose-related reduction in serum CTX levels and an increase in serum osteocalcin levels suggest a stimulatory effect of GLP-2 on bone formation [105]. In subsequent studies, nocturnal bone resorption decreased after daily administration of GLP-2 at bedtime, an effect that persisted for 2 weeks [100] and for 4 months [106]. In this double-blind placebocontrolled study 160 postmenopausal women were randomized to receive daily doses of 0.4, 1.6, and 3.2 mg of GLP-2 or placebo for 120 days. GLP-2 treatment resulted in a sustained decrease in bone resorption (in the presence of unaffected bone formation) and, most importantly, resulted in a dose-dependent increase in hip BMD for women randomized to the higher doses of daily GLP-2 [106]. These experimental studies indicate that incretins have a beneficial effect on bone mass and protective effects on bone quality. Therefore, it is conceivable that antidiabetic therapies which increase GIP and GLP hormone levels either as GLP-1 receptor agonists or inhibitors of DPP-4 might exert beneficial effects on bone [91]. From a clinical perspective incretin-based therapies seem to be an ideal treatment alternative in diabetics with high risk of falls and fractures due to their bone sparing potential, and most importantly, due to their low risk of hypoglycemia.

E

416 417

5

Please cite this article as: Meier C, et al, Effects of diabetes drugs on the skeleton, Bone (2015), http://dx.doi.org/10.1016/j.bone.2015.04.026

480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543

566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605 606

C

564 565

E

562 563

R

560 561

R

558 559

O

556 557

C

554 555

N

552 553

U

550 551

F

Recently, a new class of glucose-lowering agents, the sodiumglucose co-transporter 2 (SGLT2) inhibitors have become available [25,120]. Sodium-glucose co-transporters are responsible for renal glucose reabsorption with SGLT2 accounting for approximately 90% of reabsorbed glucose. SGLT2 actively transports glucose across the proximal nephron, an action which is independent of insulin [121]. Inhibition of SGLT2 results in increasing urinary excretion of glucose, thereby decreasing serum HbA1c levels to a similar extent as most oral antidiabetic drugs. Treatment with SGLT2-inhibitors (canagliflozin, dapagliflozin) is associated with modest weight loss and a decrease in blood pressure; however, genital mycotic infections and volume depletion resulting from its diuretic effect are reported side-effects [25]. Due to their mechanism of action SGLT2-inhibitors may alter calcium and phosphate homeostasis and potentially affect bone mass and fracture risk. Preliminary clinical data have shown no changes in serum calcium or 25OH-vitamin D levels, but indicate that serum phosphate, magnesium and PTH levels are increased during dapagliflozin treatment as compared to placebo [122,123]. To further elucidate the role of SGLT2-inhibitors on bone metabolism, Ljunggren et al. performed a double-blind, placebo-controlled study in adults with type 2 diabetes whose glycemia was inadequately controlled with metformin [124]. The study confirmed small increases in serum phosphate and magnesium levels from baseline, but found no changes in serum calcium or 25OH-vitamin D and PTH levels. In addition and compared with placebo, no significant changes in bone turnover markers (P1NP, CTX) and BMD were observed during the course of 50 weeks of dapagliflozin treatment [124]. These data, however, are in contrast with recently published skeletal safety data with increased fracture incidence associated with the use of dapagliflozin [125] and canagliflozin [126]. In a double-blind, placebocontrolled study in which the long-term effects of dapagliflozin on glycemic control in 252 patients with inadequately controlled type 2 diabetes and moderate renal impairment were evaluated, only patients in the dapagliflozin groups experienced a low-trauma fracture (n = 13, 7.7%, follow-up 104 weeks). Seven of 13 patients who sustained fracture had a history of diabetic neuropathy or exhibited orthostatic hypotension [125]. An increase in fractures by about 30% was observed in patients receiving canagliflozin in a pooled analysis of eight clinical trials (mean duration 68 weeks) [126]. Potential mechanisms of a detrimental effect of SGLT2 inhibitors on bone metabolism have recently brought forward by Taylor et al. [127]. By blocking the sodium-glucose co-transporter 2 in the proximal tubule epithelial cell SGLT2 inhibitors may reduce sodium transport which increases the availability of sodium to drive cotransport of phosphate and sodium. Increased serum phosphate is likely to provoke secretion of PTH by parathyroid glands thereby enhancing bone resorption. Furthermore, serum PTH increases the secretion of FGF23 by osteocytes which has been associated with bone disease [128]. The exact mechanisms for increased magnesium levels in patients on SGLT2-inhibitors remain unclear. Consistent with the above mentioned hypothesis, data from the FDA report on canagliflozin indicate that canagliflozin increases bone turnover as suggested by increased levels of biochemical markers of bone resorption and formation [126]. In addition, a decrease in BMD, both at the lumbar spine and the hip was detected after 52 weeks of therapy with canagliflozin [126,127]. As these data are preliminary, further studies are needed to elucidate the effect of SGLT2 inhibitors on bone metabolism and, specifically, to clarify whether such a detrimental effect follows a drug class-effect or whether it is compound-specific.

548 549

O

547

R O

SGLT-2 inhibitors

[1] Vestergaard P. Discrepancies in bone mineral density and fracture risk in patients with type 1 and type 2 diabetes—a meta-analysis. Osteoporos Int 2007;18:427–44. [2] Janghorbani M, Van Dam RM, Willett WC, Hu FB. Systematic review of type 1 and type 2 diabetes mellitus and risk of fracture. Am J Epidemiol 2007;166:495–505. [3] Schwartz AV, Vittinghoff E, Bauer DC, Hillier TA, Strotmeyer ES, Ensrud KE, et al. Association of BMD and FRAX score with risk of fracture in older adults with type 2 diabetes. JAMA 2011;305:2184–92. [4] Leslie WD, Rubin MR, Schwartz AV, Kanis JA. Type 2 diabetes and bone. J Bone Miner Res 2012;27:2231–7. [5] Nyomba BL, Verhaeghe J, Thomasset M, Lissens W, Bouillon R. Bone mineral homeostasis in spontaneously diabetic BB rats. I. Abnormal vitamin D metabolism and impaired active intestinal calcium absorption. Endocrinology 1989;124: 565–72. [6] Balint E, Szabo P, Marshall CF, Sprague SM. Glucose-induced inhibition of in vitro bone mineralization. Bone 2001;28:21–8. [7] Saito M, Fujii K, Mori Y, Marumo K. Role of collagen enzymatic and glycation induced cross-links as a determinant of bone quality in spontaneously diabetic WBN/Kob rats. Osteoporos Int 2006;17:1514–23. [8] Vashishth D, Gibson GJ, Khoury JI, Schaffler MB, Kimura J, Fyhrie DP. Influence of nonenzymatic glycation on biomechanical properties of cortical bone. Bone 2001; 28:195–201. [9] Yamamoto M, Yamaguchi T, Yamauchi M, Sugimoto T. Low serum level of the endogenous secretory receptor for advanced glycation end products (esRAGE) is a risk factor for prevalent vertebral fractures independent of bone mineral density in patients with type 2 diabetes. Diabetes Care 2009;32:2263–8. [10] Schwartz AV, Garnero P, Hillier TA, Sellmeyer DE, Strotmeyer ES, Feingold KR, et al. Pentosidine and increased fracture risk in older adults with type 2 diabetes. J Clin Endocrinol Metab 2009;94:2380–6. [11] Starup-Linde J, Vestergaard P. Biochemical bone turnover markers in diabetes mellitus—a systematic review. Bone 2015. [12] Krakauer JC, McKenna MJ, Buderer NF, Rao DS, Whitehouse FW, Parfitt AM. Bone loss and bone turnover in diabetes. Diabetes 1995;44:775–82. [13] Kume S, Kato S, Yamagishi S, Inagaki Y, Ueda S, Arima N, et al. Advanced glycation end-products attenuate human mesenchymal stem cells and prevent cognate differentiation into adipose tissue, cartilage, and bone. J Bone Miner Res 2005;20: 1647–58. [14] Sanguineti R, Storace D, Monacelli F, Federici A, Odetti P. Pentosidine effects on human osteoblasts in vitro. Ann N Y Acad Sci 2008;1126:166–72. [15] McCarthy AD, Uemura T, Etcheverry SB, Cortizo AM. Advanced glycation endproducts interefere with integrin-mediated osteoblastic attachment to a type-I collagen matrix. Int J Biochem Cell Biol 2004;36:840–8. [16] Gennari L, Merlotti D, Valenti R, Ceccarelli E, Ruvio M, Pietrini MG, et al. Circulating sclerostin levels and bone turnover in type 1 and type 2 diabetes. J Clin Endocrinol Metab 2012;97:1737–44. [17] Gaudio A, Privitera F, Battaglia K, Torrisi V, Sidoti MH, Pulvirenti I, et al. Sclerostin levels associated with inhibition of the Wnt/beta-catenin signaling and reduced bone turnover in type 2 diabetes mellitus. J Clin Endocrinol Metab 2012;97: 3744–50. [18] Heilmeier U, Carpenter DR, Patsch JM, Harnish R, Joseph GB, Burghardt AJ, et al. Volumetric femoral BMD, bone geometry, and serum sclerostin levels differ between type 2 diabetic postmenopausal women with and without fragility fractures. Osteoporos Int 2015;26:1283–93. [19] Wang A, Midura RJ, Vasanji A, Wang AJ, Hascall VC. Hyperglycemia diverts dividing osteoblastic precursor cells to an adipogenic pathway and induces synthesis of a hyaluronan matrix that is adhesive for monocytes. J Biol Chem 2014;289: 11410–20. [20] Li CI, Liu CS, Lin WY, Chen CC, Yang SY, Chen HJ, et al. Glycated hemoglobin level and risk of hip fracture in older people with type 2 diabetes: a competing risk analysis of Taiwan Diabetes Cohort Study. J Bone Miner Res 2015. [21] Nicodemus KK, Folsom AR. Type 1 and type 2 diabetes and incident hip fractures in postmenopausal women. Diabetes Care 2001;24:1192–7. [22] Oei L, Zillikens MC, Dehghan A, Buitendijk GH, Castano-Betancourt MC, Estrada K, et al. High bone mineral density and fracture risk in type 2 diabetes as skeletal complications of inadequate glucose control: the Rotterdam Study. Diabetes Care 2013;36:1619–28. [23] Schneider AL, Williams EK, Brancati FL, Blecker S, Coresh J, Selvin E. Diabetes and risk of fracture-related hospitalization: the Atherosclerosis Risk in Communities Study. Diabetes Care 2013;36:1153–8. [24] Schwartz AV, Vittinghoff E, Margolis KL, Scibora LM, Palermo L, Ambrosius WT, et al. Intensive glycemic control and thiazolidinedione use: effects on cortical and trabecular bone at the radius and tibia. Calcif Tissue Int 2013;92:477–86. [25] Inzucchi SE, Bergenstal RM, Buse JB, Diamant M, Ferrannini E, Nauck M, et al. Management of hyperglycemia in type 2 diabetes, 2015: a patient-centered approach: update to a position statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 2015;38: 140–9. [26] Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest 2001;108:1167–74. [27] Jang WG, Kim EJ, Bae IH, Lee KN, Kim YD, Kim DK, et al. Metformin induces osteoblast differentiation via orphan nuclear receptor SHP-mediated transactivation of Runx2. Bone 2011;48:885–93. [28] Mai QG, Zhang ZM, Xu S, Lu M, Zhou RP, Zhao L, et al. Metformin stimulates osteoprotegerin and reduces RANKL expression in osteoblasts and ovariectomized rats. J Cell Biochem 2011;112:2902–9.

T

546

607

References

P

beneficial effect of GLP- receptor analogs and DPP-4 inhibitors on bone metabolism.

D

544 545

C. Meier et al. / Bone xxx (2015) xxx–xxx

E

6

Please cite this article as: Meier C, et al, Effects of diabetes drugs on the skeleton, Bone (2015), http://dx.doi.org/10.1016/j.bone.2015.04.026

608 609 610 611 612 613 614 615 616 617 618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 Q4 638 639 640 641 642 643 644 645 646 647 648 649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 Q5 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691

C. Meier et al. / Bone xxx (2015) xxx–xxx

N C O

R

R

E

C

D

P

R O

O

F

[58] Bone HG, Lindsay R, McClung MR, Perez AT, Raanan MG, Spanheimer RG. Effects of pioglitazone on bone in postmenopausal women with impaired fasting glucose or impaired glucose tolerance: a randomized, double-blind, placebo-controlled study. J Clin Endocrinol Metab 2013;98:4691–701. [59] Gruntmanis U, Fordan S, Ghayee HK, Abdullah SM, See R, Ayers CR, et al. The peroxisome proliferator-activated receptor-gamma agonist rosiglitazone increases bone resorption in women with type 2 diabetes: a randomized, controlled trial. Calcif Tissue Int 2010;86:343–9. [60] Grey A, Bolland M, Gamble G, Wattie D, Horne A, Davidson J, et al. The peroxisomeproliferator-activated receptor-gamma agonist rosiglitazone decreases bone formation and bone mineral density in healthy postmenopausal women: a randomized, controlled trial. J Clin Endocrinol Metab 2007;92:1305–10. [61] van Lierop AH, Hamdy NA, van der Meer RW, Jonker JT, Lamb HJ, Rijzewijk LJ, et al. Distinct effects of pioglitazone and metformin on circulating sclerostin and biochemical markers of bone turnover in men with type 2 diabetes mellitus. Eur J Endocrinol 2012;166:711–6. [62] Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, Downes M, et al. PPARgamma signaling and metabolism: the good, the bad and the future. Nat Med 2013;19: 557–66. [63] Lecka-Czernik B, Moerman EJ, Grant DF, Lehmann JM, Manolagas SC, Jilka RL. Divergent effects of selective peroxisome proliferator-activated receptor-gamma 2 ligands on adipocyte versus osteoblast differentiation. Endocrinology 2002;143: 2376–84. [64] Shockley KR, Lazarenko OP, Czernik PJ, Rosen CJ, Churchill GA, Lecka-Czernik B. PPARγ2 nuclear receptor controls multiple regulatory pathways of osteoblast differentiation from marrow mesenchymal stem cells. J Cell Biochem 2009;106: 232–46. [65] Lazarenko OP, Rzonca SO, Hogue WR, Swain FL, Suva LJ, Lecka-Czernik B. Rosiglitazone induces decreases in bone mass and strength that are reminiscent of aged bone. Endocrinology 2007. [66] Wan Y, Chong LW, Evans RM. PPAR-gamma regulates osteoclastogenesis in mice. Nat Med 2007;13:1496–503. [67] Wei W, Wang X, Yang M, Smith LC, Dechow PC, Wan Y. PGC1beta mediates PPARgamma activation of osteoclastogenesis and rosiglitazone-induced bone loss. Cell Metab 2010;11:503–16. [68] Sun H, Kim JK, Mortensen R, Mutyaba LP, Hankenson KD, Krebsbach PH. Osteoblast-targeted suppression of PPARgamma increases osteogenesis through activation of mTOR signaling. Stem Cells 2013;31:2183–92. [69] Rzonca SO, Suva LJ, Gaddy D, Montague DC, Lecka-Czernik B. Bone is a target for the antidiabetic compound rosiglitazone. Endocrinology 2004;145:401–6. [70] Sottile V, Seuwen K, Kneissel M. Enhanced marrow adipogenesis and bone resorption in estrogen-deprived rats treated with the PPARgamma agonist BRL49653 (rosiglitazone). Calcif Tissue Int 2004;75:329–37. [71] Soroceanu MA, Miao D, Bai XY, Su H, Goltzman D, Karaplis AC. Rosiglitazone impacts negatively on bone by promoting osteoblast/osteocyte apoptosis. J Endocrinol 2004;183:203–16. [72] Cock TA, Back J, Elefteriou F, Karsenty G, Kastner P, Chan S, et al. Enhanced bone formation in lipodystrophic PPARgamma(hyp/hyp) mice relocates haematopoiesis to the spleen. EMBO Rep 2004;5:1007–12. [73] Ali AA, Weinstein RS, Stewart SA, Parfitt AM, Manolagas SC, Jilka RL. Rosiglitazone causes bone loss in mice by suppressing osteoblast differentiation and bone formation. Endocrinology 2005;146:1226–35. [74] Akune T, Ohba S, Kamekura S, Yamaguchi M, Chung UI, Kubota N, et al. PPARgamma insufficiency enhances osteogenesis through osteoblast formation from bone marrow progenitors. J Clin Invest 2004;113:846–55. [75] Cao J, Ou G, Yang N, Ding K, Kream BE, Hamrick MW, et al. Impact of targeted PPARgamma disruption on bone remodeling. Mol Cell Endocrinol 2015. [76] Lecka-Czernik B, Ackert-Bicknell C, Adamo ML, Marmolejos V, Churchill GA, Shockley KR, et al. Activation of peroxisome proliferator-activated receptor gamma (PPARgamma) by rosiglitazone suppresses components of the insulinlike growth factor regulatory system in vitro and in vivo. Endocrinology 2007; 148:903–11. [77] Moerman EJ, Teng K, Lipschitz DA, Lecka-Czernik B. Aging activates adipogenic and suppresses osteogenic programs in mesenchymal marrow stroma/stem cells: the role of PPAR-gamma2 transcription factor and TGF-beta/BMP signaling pathways. Aging Cell 2004;3:379–89. [78] Tontonoz P, Spiegelman BM. Fat and beyond: the diverse biology of PPARgamma. Annu Rev Biochem 2008;77:289–312. [79] Choi JH, Banks AS, Kamenecka TM, Busby SA, Chalmers MJ, Kumar N, et al. Antidiabetic actions of a non-agonist PPARgamma ligand blocking Cdk5-mediated phosphorylation. Nature 2011;477:477–81. [80] Rahman S, Czernik PJ, Lu Y, Lecka-Czernik B. Beta-catenin directly sequesters adipocytic and insulin sensitizing activities but not osteoblastic activity of PPARgamma2 in marrow mesenchymal stem cells. PLoS ONE 2012;7:e51746. [81] Hinds Jr TD, Stechschulte LA, Cash HA, Whisler D, Banerjee A, Yong W, et al. Protein phosphatase 5 mediates lipid metabolism through reciprocal control of glucocorticoid receptor and peroxisome proliferator-activated receptor-gamma (PPARgamma). J Biol Chem 2011;286:42911–22. [82] Stechschulte LA, Hinds Jr TD, Ghanem SS, Shou W, Najjar SM, Sanchez ER. FKBP51 reciprocally regulates GRalpha and PPARgamma activation via the Akt-p38 pathway. Mol Endocrinol 2014;28:1254–64. [83] Higgins LS, Depaoli AM. Selective peroxisome proliferator-activated receptor gamma (PPARgamma) modulation as a strategy for safer therapeutic PPARgamma activation. Am J Clin Nutr 2010;91:267S–72S. [84] Lazarenko OP, Rzonca SO, Suva LJ, Lecka-Czernik B. Netoglitazone is a PPAR-gamma ligand with selective effects on bone and fat. Bone 2006;38:74–85.

E

T

[29] Sedlinsky C, Molinuevo MS, Cortizo AM, Tolosa MJ, Felice JI, Sbaraglini ML, et al. Metformin prevents anti-osteogenic in vivo and ex vivo effects of rosiglitazone in rats. Eur J Pharmacol 2011;668:477–85. [30] Kahn SE, Zinman B, Lachin JM, Haffner SM, Herman WH, Holman RR, et al. Rosiglitazone-associated fractures in type 2 diabetes: an Analysis from a Diabetes Outcome Progression Trial (ADOPT). Diabetes Care 2008;31:845–51. [31] Zinman B, Haffner SM, Herman WH, Holman RR, Lachin JM, Kravitz BG, et al. Effect of rosiglitazone, metformin, and glyburide on bone biomarkers in patients with type 2 diabetes. J Clin Endocrinol Metab 2010;95:134–42. [32] Melton III LJ, Leibson CL, Achenbach SJ, Therneau TM, Khosla S. Fracture risk in type 2 diabetes: update of a population-based study. J Bone Miner Res 2008;23: 1334–42. [33] Vestergaard P, Rejnmark L, Mosekilde L. Diabetes and its complications and their relationship with risk of fractures in type 1 and 2 diabetes. Calcif Tissue Int 2009; 84:45–55. [34] Meier C, Kraenzlin ME, Bodmer M, Jick SS, Jick H, Meier CR. Use of thiazolidinediones and fracture risk. Arch Intern Med 2008;168:820–5. [35] Monami M, Cresci B, Colombini A, Pala L, Balzi D, Gori F, et al. Bone fractures and hypoglycemic treatment in type 2 diabetic patients: a case–control study. Diabetes Care 2008;31:199–203. [36] Kanazawa I, Yamaguchi T, Yamamoto M, Sugimoto T. Relationship between treatments with insulin and oral hypoglycemic agents versus the presence of vertebral fractures in type 2 diabetes mellitus. J Bone Miner Metab 2010;28:554–60. [37] Bilik D, McEwen LN, Brown MB, Pomeroy NE, Kim C, Asao K, et al. Thiazolidinediones and fractures: evidence from translating research into action for diabetes. J Clin Endocrinol Metab 2010;95:4560–5. [38] Colhoun HM, Livingstone SJ, Looker HC, Morris AD, Wild SH, Lindsay RS, et al. Scottish Diabetes Research Network Epidemiology G. Hospitalised hip fracture risk with rosiglitazone and pioglitazone use compared with other glucoselowering drugs. Diabetologia 2012;55:2929–37. [39] Napoli N, Strotmeyer ES, Ensrud KE, Sellmeyer DE, Bauer DC, Hoffman AR, et al. Fracture risk in diabetic elderly men: the MrOS study. Diabetologia 2014;57: 2057–65. [40] Tzoulaki I, Molokhia M, Curcin V, Little MP, Millett CJ, Ng A, et al. Risk of cardiovascular disease and all cause mortality among patients with type 2 diabetes prescribed oral antidiabetes drugs: retrospective cohort study using UK general practice research database. BMJ 2009;339:b4731. [41] Kahn SE, Haffner SM, Heise MA, Herman WH, Holman RR, Jones NP, et al. Glycemic durability of rosiglitazone, metformin, or glyburide monotherapy. N Engl J Med 2006;355:2427–43. [42] Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med 2007;356:2457–71. [43] Lincoff AM, Wolski K, Nicholls SJ, Nissen SE. Pioglitazone and risk of cardiovascular events in patients with type 2 diabetes mellitus: a meta-analysis of randomized trials. JAMA 2007;298:1180–8. [44] Yki-Jarvinen H. Thiazolidinediones. N Engl J Med 2004;351:1106–18. [45] Grey A. Skeletal consequences of thiazolidinedione therapy. Osteoporos Int 2008; 19:129–37. [46] Lecka-Czernik B. Bone loss in diabetes: use of antidiabetic thiazolidinediones and secondary osteoporosis. Curr Osteoporos Rep 2010;8:178–84. [47] Takeda. Observation of an increased incidence of fractures in female patients who received long-term treatment with Actos (pioglitazone HCl) tablets for type 2 diabetes mellitus. Letter to health care providers. Takeda Pharmaceuticals North America; 2007. [48] Loke YK, Singh S, Furberg CD. Long-term use of thiazolidinediones and fractures in type 2 diabetes: a meta-analysis. CMAJ 2009;180:32–9. [49] Zhu ZN, Jiang YF, Ding T. Risk of fracture with thiazolidinediones: an updated metaanalysis of randomized clinical trials. Bone 2014;68:115–23. [50] Douglas IJ, Evans SJ, Pocock S, Smeeth L. The risk of fractures associated with thiazolidinediones: a self-controlled case-series study. PLoS Med 2009;6: e1000154. [51] Grey A, Bolland M, Gamble G, Wattie D, Horne A, Davidson J, et al. The peroxisome proliferator-activated receptor-{gamma} agonist rosiglitazone decreases bone formation and bone mineral density in healthy postmenopausal women: a randomized, controlled trial. J Clin Endocrinol Metab 2007;92:1305–10. [52] Glintborg D, Andersen M, Hagen C, Heickendorff L, Hermann AP. Association of pioglitazone treatment with decreased bone mineral density in obese premenopausal patients with polycystic ovary syndrome: a randomized, placebocontrolled trial. J Clin Endocrinol Metab 2008;93:1696–701. [53] Berberoglu Z, Yazici AC, Demirag NG. Effects of rosiglitazone on bone mineral density and remodelling parameters in postmenopausal diabetic women: a 2-year follow-up study. Clin Endocrinol (Oxf) 2010;73:305–12. [54] Borges JL, Bilezikian JP, Jones-Leone AR, Acusta AP, Ambery PD, Nino AJ, et al. A randomized, parallel group, double-blind, multicentre study comparing the efficacy and safety of Avandamet (rosiglitazone/metformin) and metformin on long-term glycaemic control and bone mineral density after 80 weeks of treatment in drugnaive type 2 diabetes mellitus patients. Diabetes Obes Metab 2011;13:1036–46. [55] Bilezikian JP, Josse RG, Eastell R, Lewiecki EM, Miller CG, Wooddell M, et al. Rosiglitazone decreases bone mineral density and increases bone turnover in postmenopausal women with type 2 diabetes mellitus. J Clin Endocrinol Metab 2013. [56] Grey A, Bolland M, Fenwick S, Horne A, Gamble G, Drury PL, et al. The skeletal effects of pioglitazone in type 2 diabetes or impaired glucose tolerance: a randomized controlled trial. Eur J Endocrinol 2014;170:255–62. [57] Harslof T, Wamberg L, Moller L, Stodkilde-Jorgensen H, Ringgaard S, Pedersen SB, et al. Rosiglitazone decreases bone mass and bone marrow fat. J Clin Endocrinol Metab 2011;96:1541–8.

U

692 693 694 695 696 697 698 Q6 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715 716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 Q7 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 Q8 772 773 774 775 776 777

7

Please cite this article as: Meier C, et al, Effects of diabetes drugs on the skeleton, Bone (2015), http://dx.doi.org/10.1016/j.bone.2015.04.026

778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 Q9 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 Q10 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863

[113] [114]

[115]

[116]

[117]

[118] [119]

D

[120]

[121]

[122]

C

E

R

R

[123] [124]

[125]

[126]

[127] [128]

N

C

O

F

[112]

O

[111]

R O

[110]

suppressing bone resorption in aged ovariectomized rats. J Bone Miner Res 2013; 28:1641–52. Kim JY, Lee SK, Jo KJ, Song DY, Lim DM, Park KY, et al. Exendin-4 increases bone mineral density in type 2 diabetic OLETF rats potentially through the downregulation of SOST/sclerostin in osteocytes. Life Sci 2013;92:533–40. Bunck MC, Eliasson B, Corner A, Heine RJ, Shaginian RM, Taskinen MR, et al. Exenatide treatment did not affect bone mineral density despite body weight reduction in patients with type 2 diabetes. Diabetes Obes Metab 2011;13:374–7. Mabilleau G, Mieczkowska A, Chappard D. Use of glucagon-like peptide-1 receptor agonists and bone fractures: a meta-analysis of randomized clinical trials. J Diabetes 2014;6:260–6. Su B, Sheng H, Zhang M, Bu L, Yang P, Li L, et al. Risk of bone fractures associated with glucagon-like peptide-1 receptor agonists' treatment: a meta-analysis of randomized controlled trials. Endocrine 2015;48:107–15. Mabilleau G. Use of GLP-1 mimetic in type 2 diabetes mellitus: is it the end of fragility fractures? Endocrine 2015;48:1–2. Glorie L, Behets GJ, Baerts L, De Meester I, D'Haese PC, Verhulst A. DPP4 inhibitor treatment attenuates bone loss and improves mechanical bone strength in male diabetic rats. Am J Physiol Endocrinol Metab 2014. Bunck MC, Poelma M, Eekhoff EM, Schweizer A, Heine RJ, Nijpels G, et al. Effects of vildagliptin on postprandial markers of bone resorption and calcium homeostasis in recently diagnosed, well-controlled type 2 diabetes patients. J Diabetes 2012; 4:181–5. Monami M, Dicembrini I, Antenore A, Mannucci E. Dipeptidyl peptidase-4 inhibitors and bone fractures: a meta-analysis of randomized clinical trials. Diabetes Care 2011;34:2474–6. Driessen JH, van Onzenoort HA, Henry RM, Lalmohamed A, van den Bergh JP, Neef C, et al. Use of dipeptidyl peptidase-4 inhibitors for type 2 diabetes mellitus and risk of fracture. Bone 2014;68:124–30. Montagnani A, Gonnelli S. Antidiabetic therapy effects on bone metabolism and fracture risk. Diabetes Obes Metab 2013;15:784–91. Scirica BM, Bhatt DL, Braunwald E, Steg PG, Davidson J, Hirshberg B, et al. Investigators. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med 2013;369:1317–26. Vasilakou D, Karagiannis T, Athanasiadou E, Mainou M, Liakos A, Bekiari E, et al. Sodium-glucose cotransporter 2 inhibitors for type 2 diabetes: a systematic review and meta-analysis. Ann Intern Med 2013;159:262–74. Chao EC, Henry RR. SGLT2 inhibition—a novel strategy for diabetes treatment. Nat Rev Drug Discov 2010;9:551–9. Nauck MA, Del Prato S, Meier JJ, Duran-Garcia S, Rohwedder K, Elze M, et al. Dapagliflozin versus glipizide as add-on therapy in patients with type 2 diabetes who have inadequate glycemic control with metformin: a randomized, 52-week, double-blind, active-controlled noninferiority trial. Diabetes Care 2011;34: 2015–22. List JF, Woo V, Morales E, Tang W, Fiedorek FT. Sodium-glucose cotransport inhibition with dapagliflozin in type 2 diabetes. Diabetes Care 2009;32:650–7. Ljunggren O, Bolinder J, Johansson L, Wilding J, Langkilde AM, Sjostrom CD, et al. Dapagliflozin has no effect on markers of bone formation and resorption or bone mineral density in patients with inadequately controlled type 2 diabetes mellitus on metformin. Diabetes Obes Metab 2012;14:990–9. Kohan DE, Fioretto P, Tang W, List JF. Long-term study of patients with type 2 diabetes and moderate renal impairment shows that dapagliflozin reduces weight and blood pressure but does not improve glycemic control. Kidney Int 2014;85:962–71. Kwohn H. Canagliflozin: clinical efficacy and safety. Endocrinology and Metabolic Drugs Advisory Committee Meeting; 2013 [www.fda.gov/downloads/AdvisoryCommitteesMeetingMaterials/Drugs/ EndocrinologicandMetabolicDrugsAdvisoryCommittee/UCM336234.pdf. In.]. Taylor SI, Blau JE, Rother KI. Possible adverse effects of SGLT2 inhibitors on bone. Lancet Diabetes Endocrinol 2015;3:8–10. Quarles LD. Skeletal secretion of FGF-23 regulates phosphate and vitamin D metabolism. Nat Rev Endocrinol 2012;8:276–86.

P

[109]

T

[85] Lee DH, Huang H, Choi K, Mantzoros C, Kim YB. Selective PPARgamma modulator INT131 normalizes insulin signaling defects and improves bone mass in dietinduced obese mice. Am J Physiol Endocrinol Metab 2012;302:E552–60. [86] Fukunaga T, Zou W, Rohatgi N, Colca JR, Teitelbaum SL. An insulin-sensitizing thiazolidinedione, which minimally activates PPAR, does not cause bone loss. J Bone Miner Res 2014. [87] Kolli V, Stechschulte LA, Dowling AR, Rahman S, Czernik PJ, Lecka-Czernik B. Partial agonist, telmisartan, maintains PPARgamma serine 112 phosphorylation, and does not affect osteoblast differentiation and bone mass. PLoS ONE 2014;9:e96323. [88] Choi JH, Banks AS, Estall JL, Kajimura S, Bostrom P, Laznik D, et al. Anti-diabetic drugs inhibit obesity-linked phosphorylation of PPARgamma by Cdk5. Nature 2010;466:451–6. [89] Vestergaard P, Rejnmark L, Mosekilde L. Relative fracture risk in patients with diabetes mellitus, and the impact of insulin and oral antidiabetic medication on relative fracture risk. Diabetologia 2005;48:1292–9. [90] Zinman B, Haffner SM, Herman WH, Holman RR, Lachin JM, Kravitz BG, et al. Effect of rosiglitazone, metformin, and glyburide on bone biomarkers in patients with type 2 diabetes. J Clin Endocrinol Metab 2010;95:134–42. [91] Lecka-Czernik B. Safety of anti-diabetic therapies on bone. Clin Rev Bone Miner Metab 2013;11:49–58. [92] Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). UK Prospective Diabetes Study (UKPDS) Group. Lancet 1998;352: 837–53. [93] Lapane KL, Yang S, Brown MJ, Jawahar R, Pagliasotti C, Rajpathak S. Sulfonylureas and risk of falls and fractures: a systematic review. Drugs Aging 2013;30:527–47. [94] Campbell JE, Drucker DJ. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab 2013;17:819–37. [95] Janssen P, Rotondo A, Mule F, Tack J. Review article: a comparison of glucagon-like peptides 1 and 2. Aliment Pharmacol Ther 2013;37:18–36. [96] Drucker DJ, Yusta B. Physiology and pharmacology of the enteroendocrine hormone glucagon-like peptide-2. Annu Rev Physiol 2014;76:561–83. [97] Deacon CF. Circulation and degradation of GIP and GLP-1. Horm Metab Res 2004; 36:761–5. [98] Kieffer TJ, Habener JF. The glucagon-like peptides. Endocr Rev 1999;20:876–913. [99] Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007; 132:2131–57. [100] Henriksen DB, Alexandersen P, Hartmann B, Adrian CL, Byrjalsen I, Bone HG, et al. Disassociation of bone resorption and formation by GLP-2: a 14-day study in healthy postmenopausal women. Bone 2007;40:723–9. [101] Xie D, Zhong Q, Ding KH, Cheng H, Williams S, Correa D, et al. Glucose-dependent insulinotropic peptide-overexpressing transgenic mice have increased bone mass. Bone 2007;40:1352–60. [102] Zhong Q, Itokawa T, Sridhar S, Ding KH, Xie D, Kang B, et al. Effects of glucosedependent insulinotropic peptide on osteoclast function. Am J Physiol Endocrinol Metab 2007;292:E543–8. [103] Yamada C, Yamada Y, Tsukiyama K, Yamada K, Udagawa N, Takahashi N, et al. The murine glucagon-like peptide-1 receptor is essential for control of bone resorption. Endocrinology 2008;149:574–9. [104] Walsh JS, Henriksen DB. Feeding and bone. Arch Biochem Biophys 2010;503:11–9. [105] Henriksen DB, Alexandersen P, Byrjalsen I, Hartmann B, Bone HG, Christiansen C, et al. Reduction of nocturnal rise in bone resorption by subcutaneous GLP-2. Bone 2004;34:140–7. [106] Henriksen DB, Alexandersen P, Hartmann B, Adrian CL, Byrjalsen I, Bone HG, et al. Four-month treatment with GLP-2 significantly increases hip BMD: a randomized, placebo-controlled, dose-ranging study in postmenopausal women with low BMD. Bone 2009;45:833–42. [107] Nielsen LL, Baron AD. Pharmacology of exenatide (synthetic exendin-4) for the treatment of type 2 diabetes. Curr Opin Investig Drugs 2003;4:401–5. [108] Ma X, Meng J, Jia M, Bi L, Zhou Y, Wang Y, et al. Exendin-4, a glucagon-like peptide1 receptor agonist, prevents osteopenia by promoting bone formation and

U

864 865 866 867 868 869 Q11 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887 888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924

C. Meier et al. / Bone xxx (2015) xxx–xxx

E

8

Please cite this article as: Meier C, et al, Effects of diabetes drugs on the skeleton, Bone (2015), http://dx.doi.org/10.1016/j.bone.2015.04.026

925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 Q12 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973 974 975 976 977 978

Effects of diabetes drugs on the skeleton.

Type 2 diabetes is associated with increased fracture risk and the mechanisms underlying the detrimental effects of diabetes on skeletal health are on...
361KB Sizes 0 Downloads 9 Views