Skip to main content
Erschienen in: BMC Nephrology 1/2018

Open Access 01.12.2018 | Research article

Outcomes of bisphosphonate and its supplements for bone loss in kidney transplant recipients: a systematic review and network meta-analysis

verfasst von: Yan Yang, Shi Qiu, Linghui Deng, Xi Tang, Xinrui Li, Qiang Wei, Ping Fu

Erschienen in: BMC Nephrology | Ausgabe 1/2018

Abstract

Background

Mineral bone disease constitutes a common complication of post-kidney transplantation, leading to great disability. As there is no consensus on the optimal treatment for post-kidney transplant recipients (KTRs), we aimed to evaluate the efficacy and safety of bisphosphonate and its combined therapies.

Methods

We incorporated relevant trials to perform a network meta-analysis from direct and indirect comparisons. We searched PubMed, Embase and the CENTRAL and the reference lists of relevant articles up to August 1, 2017, for randomized controlled trials. The primary outcome was bone mineral density (BMD) change at the femoral neck and the lumbar spine.

Results

From a total of 864 citations, 18 randomized controlled trials with a total of 1200 participants were included. Five different regimens were considered. Bisphosphonate plus calcium revealed a significant gain in percent BMD change than calcium alone at the femoral neck (mean difference (MD), 5.83; 95% credible interval (CrI), 1.61 to 9.27). No significant difference was detected when restricting to absolute terms. At the lumbar spine, bisphosphonate and calcium with or without vitamin D analogs outperformed calcium solely (MD, 0.07; 95% CrI, 0.00 to 0.13; MD, 0.06; 95% CrI, 0.02 to 0.09). Compared to calcium with vitamin D analogs, adding bisphosphonate was associated with marked improvement (MD, 0.03; 95% CrI, 0.00 to 0.05). Considering percent terms, combination of bisphosphonate with calcium and vitamin D analogs showed greater beneficial effects than calcium alone or with either vitamin D analogs or calcitonin (MD, 10.51; 95% CrI, 5.92 to 15.34; MD, 5.48; 95% CrI, 2.57 to 8.42; MD, 6.39; 95% CrI, 0.55 to 12.89). Both bisphosphonate and vitamin D analogs combined with calcium displayed a notable improvement compared to calcium alone (MD, 7.24; 95% CrI, 3.73 to 10.69; MD, 5.02; 95% CrI, 1.20 to 8.84).

Conclusions

Our study suggested that additional use of bisphosphonate was well-tolerated and more favorable in KTRs to improve BMD.

Background

Since kidney transplantation (KT) became an effective treatment of patients with end-stage renal disease (ESRD), clinicians have paid more attention to complications of kidney transplant recipients (KTRs). Post-transplantation bone disease which can result in serious disabilities and fractures has been observed among a large proportion of KTRs [1]. According to Naylor and colleagues [2], the 5-year cumulative incidence of fracture ranged from 0.85 to 27% after KT. Hence, prevention and treatment of bone disorders are of great importance to improve high-quality long-term survival of KTRs.
The etiology of transplant bone disease is multifactorial and most KTRs have preexisting chronic kidney disease-mineral and bone disorders (CKD-MBD) [3]. Apart from these, glucocorticoid-induced suppression of bone formation, calcineurin inhibitors (CNIs) and persistent hyperparathyroidism are the most important risk factors for bone loss [46]. Postmenopausal status, prolonged immobilization, duration of CKD stage 5, smoking and presence of diabetes may also contribute to bone loss [4]. The Kidney Disease Improving Global Outcomes (KDIGO) guideline [7] suggested that “vitamin D, calcitriol/alfacalcidol, or bisphosphonates be considered for low BMD patients with stable graft function”, but it was derived from the very low quality of evidence.
Previous meta-analyses [8, 9] have demonstrated that bisphosphonates have favorable efficacy on bone mineral density (BMD), but questionable effect on the fracture risk. However, these studies did not examine the effect of co-intervention with calcium and/or vitamin D. Moreover, it is still uncertain that the optimal approach to prevent bone loss and whether it is need to use combined therapy. To obtain a better understanding on this issue, we performed a network meta-analysis (NMA). In this NMA, we systematically reviewed the literature and estimated relative treatment effects for all possible comparisons including bisphosphonates and co-intervention.

Methods

Search strategy

This systematic review is performed in keeping with Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guideline [10]. A comprehensive search was conducted in PubMed, Embase and the Cochrane Library Central Register of Controlled Trials (CENTRAL) by two independent investigators up to August 1st, 2017. The full search parameters for each database was outlined in Additional file 1. Referenced articles and systematic reviews were screened to maximize inclusion of pertinent data.

Selection criteria

Only randomized controlled trials (RCTs) comparing bisphosphonate-treated and control groups of adult KTRs were included. The full-text original article with at least one interest outcome was finally involved. Two independent investigators (YY, QS) initially screened the citation titles and abstracts. Studies were excluded because of non-English text, combined transplantation. If duplicate studies from the identical authors were found, the reports were grouped together and only the publication with a complete data was used. Any discrepancies in the study inclusion were resolved by consulting the senior authors (TX).

Data extraction and quality assessment

The independent reviewers (YY, SQ) used a standardized form to extract information from each eligible study. Data regarding study-, patient- and treatment-related characteristics and outcomes were extracted simultaneously. When relevant information was unclear or needed data was unavailable, attempts were made to obtain eligible data from the first or corresponding author of such studies. We assessed the validity of the NMA through a qualitative appraisal of study designs and methods. We executed the tool recommended by the Cochrane Collaboration to evaluate the risk of bias [11].

Outcomes

The primary outcome was the BMD change (percent change and absolute change [in g/cm2]) at the lumbar spine and the femoral neck after successful KT. The secondary outcomes were overall fractures, all-cause mortality, graft loss, acute renal rejection, adverse events. The fractures occurred during reported follow-up time that identified by radiographs were used to calculate fracture incidence. Graft loss was regarded as a doubling of the baseline serum creatinine level or progressing to ESRD again. We used data from the longest complete follow-up, when the outcomes of different follow-up intervals were reported. If investigators published more than one report addressing the same population, we included the most comprehensive report.

Data synthesis and statistical analysis

The pair-wise meta-analysis by the random-effects model was performed initially [12]. Results were expressed as mean difference (MD) with 95% confidence intervals (CI) for continuous outcomes (percent change and absolute change in BMD), while the odds ratio (OR) was used for dichotomous variables (fracture, all-cause mortality, graft loss, acute renal rejection, adverse events). The level of statistical significance was set at P < 0·05 and all statistical tests were two-sided. The statistical heterogeneity among studies was evaluated by the Cochran’s Q test and the I2 statistic. A P value of 0.05 or less for the Q test or an I2 greater than 50% was suggestive of substantial study heterogeneity.
We performed random-effects Bayesian network meta-analyses for indirect and mixed comparisons using Markov chain Monte Carlo methods in WinBUGS version 1.4.3 (MRC Biostatistics Unit) [13]. A Bayesian fixed-effect framework was deemed appropriate because of the limited number of studies supporting each edge in the network [13, 14]. We report the resultant effect as OR or MD with corresponding 95% credibility intervals (CrIs), which are the Bayesian analogue of 95% CIs. We estimated the relative ranking probability of each strategy and obtained the hierarchy of competing interventions using rankograms and surface under the cumulative ranking curve (SUCRA) [15]. The SUCRA index ranges between 0 (or 0%) and 1 (or 100%), where the treatments with highest and lowest SUCRA are considered to be the best and worst treatments, respectively.
To assess the presence of the inconsistency, we employed the node-splitting method, excluding one direct comparison at a time and estimating the indirect treatment effect for the excluded comparison. To check the assumption of consistency in the entire network, the design-by-treatment model was conducted [14]. If the total residual deviance and the effective number of parameters (pD) are almost the same, the network consistency is considered to be satisfied. We then performed sensitivity analysis and meta-regressions to explore important network inconsistency.

Quality of evidence

The quality of evidence was rated according to the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) methodology [16]. In this approach, direct evidence from RCTs starts at high quality and can be downgraded based on the risk of bias, indirectness, imprecision, inconsistency (or heterogeneity) and publication bias to levels of moderate, low and relatively low quality [17].

Results

Study characteristics

The PRISMA [10] flowchart depicting the electronic searching process is presented in Fig. 1. There are 864 potentially relevant articles identified through electronic and reference searches. According to title and abstract, 821 publications were excluded after the initial screening. A further 26 studies were excluded because they were not RCT, without available data of interest outcomes and lack of full-text. Overall, 18 RCTs (19 publications) [1836] involving 1200 participants were included in this NMA. The studies were published between October 1998 and March 2015. The details of the interventions, baseline characteristics of the populations, follow-up period were outlined for NMA in Table 1. Most of the RCTs included both sexes, except one study [26] only included male patients and two studies [25, 27] did not mention. The number of patients allocated to each treatment ranged from 8 to 66, whereas patient follow-up duration ranged from 6 months to 3 years after first administration.
Table 1
Study characteristics
Study
Follow-up
Country
No. of Patients
Female/Male
Intervention
Bisphosphonate Administration
N
Co-intervention
Immunosuppression
Smerud 2012 [18]
12 months
Norway
129
30/99
ibandronate
3 mg i.v. (every 3 months)
66
PO calcium 500 mg tweice daily+ calcitriol 0.25 mcg daily
corticosteroids, MMF, CsA or FK506
placebo
 
63
Coco 2012 [19]
12 months
USA
42
15/27
risedronate
35 mg p.o. (weekly)
20
PO calcitriol 0.25 μg daily (with or without calcium)
corticosteroids, MMF, FK506, rapamycin
placebo
 
22
Torregrosa 2010 [20]
12 months
Spain
101
34/67
risedronate
35 mg p.o. (weekly)
52
PO calcium 1.5 g daily + vitamin D 400 IU daily
corticosteroids, FK506 with or without MMF
no treatment
 
49
Torregrosa 2011 [21]
12 months
Spain
39
13/26
pamidronate
30 mg i.v. (between day 7 and 10 after KT and 3 months post-KT)
24
PO calcium 1 g daily + cholecalciferol 800 IU daily
corticosteroids, MMF, CsA
placebo
 
15
Walsh 2009 [22]
24 months
UK
125
24/69
pamidronate
1 mg/kg i.v. (perioperatively and at month 1, 4, 8, 12)
65
PO calcium 500 mg daily + vitamin D 400 IU daily
corticosteroids, CsA
no treatment
 
60
Lan 2008 [23]
6 months
China
46
27/19
alendronate
70 mg p.o. (weekly)
23
PO calcium 800 mg daily + calcitriol 0.25 μg daily
corticosteroids, MMF, CsA
no treatment
 
23
Trabulus 2008 [24]
12 months
Turkey
64
19/40
alendronate
10 mg p.o. (daily)
13
PO calcium 1 g daily
corticosteroids, azathioprin or MMF, CsA or FK506
alfacalcidol
0.5 μg p.o. (daily)
25
alendronate + alfacalcidol
17
no treatment
 
9
Nayak 2007 [25]
6 months
India
50
NA
alendronate
35 mg p.o. (weekly)
27
PO calcium 1 g daily + vitamin D
NA
no treatment
 
23
El-Agroudy 2005 [26]
12 months
Egypt
60
0/60
alendronate
5 mg p.o. (daily)
15
PO calcium 500 mg daily
corticosteroids, CsA
alfacalcidol
0.5 μg p.o. (daily)
15
calcitonin
100 μl intranasally (p.o.d and stopped for 1 month every 3 month)
15
no treatment
 
15
Schwarz 2004 [27]
36 months
Austria
20
8/12
zoledronic acid
4 mg i.v. (week 2, month 3)
9
PO calcium 1 g daily
corticosteroids, MMF, CsA
placebo
 
10
Jeffery 2003 [28]
12 months
Canada
117
26/71
alendronate
10 mg p.o. (daily)
57
PO calcium 500 mg daily
corticosteroids, CsA, azathioprin or MMF
calcitriol
0.25 μg p.o. (daily)
60
Coco 2003 [29]
12 months
USA
72
28/31
pamidronate
60 mg i.v. (< 48 h after KT, 30 mg i.v. at months 1, 2, 3, 6)
36
calcium + calcitriol
corticosteroids, CsA or FK506
no treatment
 
36
Hass 2003 [30]
6 months
Austria
20
8/12
zoledronic acid
4 mg i.v. (week 2, month 3)
10
PO calcium 1 g daily
corticosteroids, MMF, CsA
placebo
 
10
Grotz 2001 [31]
12 months
Germany
80
24/48
ibandronate
1 mg i.v. (just before KT, 2 mg i.v. at month 3, 6, 9)
36
PO calcium 500 mg daily
corticosteroids, MMF, CsA
no treatment
 
36
Nam 2000 [32]
6 months
South Kore
50
21/29
pamidronate
30 mg i.v. (every 4 weeks)
15
PO calcium 500 mg daily
NA
calcitriol
0.5 μg p.o. (daily)
15
no treatment
 
20
Grotz 1998 [33]
12 months
Germany
46
17/29
clodronate
800 mg p.o. (daily) for 14 days, each followed by 75 days without treatment
15
PO calcium 500 mg daily
corticosteroids, CsA
calcitonin
100 IU intranasally twice a day
16
no treatment
 
15
Giannini 2001 [34]
12 months
Italy
40
13/27
alendronate
10 mg p.o. (daily)
20
PO calcium 500 mg daily + calcitriol 0.5 μg daily
corticosteroids, CsA with or without azathioprin
no treatment
 
20
Koc 2002 [35]
12 months
Turkey
35
10/25
alendronate
10 mg p.o. (daily)
8
PO calcium 1 g daily
corticosteroids, azathioprin, CsA
calcitriol
0.5 μg p.o. (daily)
8
no treatment
 
8
Torregrosa 2007 [36]
12 months
Spain
84
42/42
risedronate
35 mg p.o. (weekly)
39
PO calcium 2.5 g daily + vitamin D
corticosteroids, CsA or FK506, with or without MMF
no treatment
 
45
KT: kidney transplantation; i.v.: intravenous; p.o., PO: peros; p.o.d: per other day; N: numbers; NA: not available; MMF: mycophenolate mofetil; CsA: cyclosporine; FK506: tacrolimus; AZA: azathioprine; mTOR: mammalian target of rapamycin
As expected, most studies compared bisphosphonate with vitamin D analogs (cholecalciferol, alfacalcidol, calcitriol) or placebo. All patients in the trials included received co-intervention including calcium [24, 2628, 3033, 35], vitamin D analogs [19], or both. Bisphosphonate interventions encompassed pamidronate [21, 22, 29, 32], zoledronic acid [27, 30] and ibandronate [18, 31] that were administered intravenously, while clodronate [33], alendronate [2326, 28, 34, 35] and risedronate [19, 20, 36] were given orally.

Risk of Bias assessment result

The results from the risk of bias assessment are provided in Additional file 2. Details regarding trial methodology were unsatisfactory or incomplete for the majority of studies. Overall, there were 6 (32%) studies regarded as high risk of bias. Only 10 (53%) studies performed randomized sequence generation adequately. Furthermore, the risk of bias for concealment of treatment allocation was unclear in 10 (53%) studies. Only 4 (33%) studies explicitly reported blinding of participants and investigators, whereas the remaining studies were at high or unclear risk in this regard. The investigators attempted to blind outcome assessors in 6 (32%) studies, 3 studies did not make an effort to blind assessors, and the residual studies were unclear. When the results were summarized from at least 10 studies, the publication bias accessed via funnel plot. Comparison adjusted funnel plot showed no evidence of asymmetry (Additional file 2).

Pairwise meta-analysis

Primary results of pairwise meta-analysis (direct comparisons) are summarized in Table 2. In terms of absolute change for the longest follow-up, adding bisphosphonate was associated with a marginal improvement compared to the combination of calcium and vitamin D analogs (femoral neck: MD, 0.36; 95% CI, 0.08 to 0.64; lumbar spine: MD, 0.38; 95% CI, 0.19 to 0.57). Bisphosphonate combined with calcium was significantly better than calcium alone (femoral neck: MD, 1.30; 95% CI, 0.92 to 1.68; lumbar spine: MD, 0.51; 95% CI, 0.20 to 0.82). Treatments with calcium alone displayed significantly lower absolute change at the femoral neck than combining with vitamin D analogs or calcitonin (MD, − 0.74; 95% CI, − 1.34 to − 0.14; MD, − 0.55; 95% CI, − 1.07 to − 0.03). When measured in percent terms, additional use of vitamin D analogs or bisphosphonate was significantly better than solely calcium (femoral neck: MD 1.53; 95% CI, 0.88 to 2.18; MD 1.14; 95% CI, 0.78 to 1.51; lumbar spine: MD 2.73; 95% CI, 1.95 to 3.51; MD 1.17; 95% CI, 0.80 to 1.54). Compared to calcium and vitamin D analogs, the combination of bisphosphonate and calcium showed significant improvement (femoral neck: MD, 1.55; 95% CI, 0.76 to 2.35; lumbar spine: MD, 1.53; 95% CI, 0.79 to 2.27). Bisphosphonate with calcium and vitamin D analogs also showed a significant gain at the lumbar spine compared to calcium and vitamin D analogs (MD, 1.32; 95% CI, 1.02 to 1.62).
Table 2
Summary effect size of pairwise and network meta-analysis
Comparisons
No. of directed trials (participants)
Pairwise meta-analysis mean differences (95% CI)
Network meta-analysis mean differences (95% CrI)
Heterogeneity I2
P-Value
Quality of evidence
Absolute BMD change at the femoral neck (536)
 Bis+Ca vs. Bis+Ca + Vit D
1 (29)
− 0.01 (− 0.32, 0.29)
Low
 Bis+Ca vs. Ca
5 (167)
1.3 (0.92, 1.68)
0.19 (− 0.01, 0.38)
94.70%
0.000
Low
 Bis+Ca vs. Ca + Vit D
2 (176)
0.26 (−0.04, 0.56)
0.06 (− 0.15, 0.26)
38.10%
0.184
Moderate
 Bis+Ca vs. Ca + Calcitonin
2 (61)
0.21 (−0.29, 0.72)
0.06 (− 0.22, 0.36)
24.60%
0.249
Moderate
 Bis+Ca + Vit D vs. Ca
0.20 (−0.14, 0.53)
Very low
 Bis+Ca + Vit D vs. Ca + Vit D
4 (206)
0.36 (0.08, 0.64)
0.07 (−0.18, 0.30)
67.60%
0.026
Low
 Bis+Ca + Vit D vs. Ca + Calcitonin
0.07 (−0.34, 0.46)
Very low
 Ca vs. Ca + Vit D
2 (46)
−0.74 (−1.34, − 0.14)
−0.13 (− 0.38, 0.13)
0.00%
0.403
Low
 Ca vs. Ca + Calcitonin
2 (60)
−0.55 (−1.07, − 0.03)
−0.12 (− 0.41, 0.19)
60.20%
0.113
Low
 Ca + Vit D vs. Ca + Calcitonin
1 (30)
0.00 (−0.30, 0.34)
Low
Percent BMD change at the femoral neck (284)
 Bis+Ca vs. Bis+Ca + Vit D
1 (29)
−4.60 (−18.07, 7.67)
Low
 Bis+Ca vs. Ca
4 (152)
1.14 (0.78, 1.51)
5.83 (1.61, 9.27)
91.10%
0.000
Moderate
 Bis+Ca vs. Ca + Vit D
4 (46)
1.55 (0.76, 2.35)
−0.24 (5.62, 9.79)
96.10%
0.000
Low
 Bis+Ca vs. Ca + Calcitonin
1 (31)
−0.04 (−19.65, 18.12)
Low
 Bis+Ca + Vit D vs. Ca
10.43 (−2.64, 23.31)
Very low
 Bis+Ca + Vit D vs. Ca + Vit D
4.35 (−2.29, 11.37)
Very low
 Bis+Ca + Vit D vs. Ca + Calcitonin
4.56 (−18.36, 19.16)
Very low
 Ca vs. Ca + Vit D
3 (51)
−1.53 (−2.18, −0.88)
−6.07 (− 17.09, 4.47)
79.30%
0.028
Low
 Ca vs. Ca + Calcitonin
1 (30)
−5.87 (−20.01, 18.60)
Low
 Ca + Vit D vs. Ca + Calcitonin
1 (30)
0.20 (−19.15, 19.61)
Low
Absolute BMD change at the lumbar spine (814)
 Bis+Ca vs. Bis+Ca + Vit D
1 (29)
−0.01 (−0.06, 0.04)
Low
 Bis+Ca vs. Ca
5 (167)
0.51 (0.20, 0.82)
0.06 (0.02, 0.09)
0.00%
0.571
Moderate
 Bis+Ca vs. Ca + Vit D
4 (176)
0.19 (−0.11, 0.49)
0.01 (−0.03, 0.06)
0.00%
0.866
Moderate
 Bis+Ca vs. Ca + Calcitonin
2 (61)
0.49 (−0.02, 1.00)
0.05 (−0.01, 0.11)
24.60%
0.250
Moderate
 Bis+Ca + Vit D vs. Ca
1 (30)
0.07 (0.00, 0.13)
Low
 Bis+Ca + Vit D vs. Ca + Vit D
8 (484)
0.38 (0.19, 0.57)
0.03 (0.00, 0.05)
92.10%
0.000
Moderate
 Bis+Ca + Vit D vs. Ca + Calcitonin
0.06 (−0.01, 0.15)
Very low
 Ca vs. Ca + Vit D
2 (46)
−0.40 (− 0.99, 0.18)
−0.04 (− 0.10, 0.02)
0.00%
0.960
Moderate
 Ca vs. Ca + Calcitonin
2 (60)
−0.04 (− 0.55, 0.47)
−0.01 (− 0.07, 0.06)
0.00%
0.874
Moderate
 Ca + Vit D vs. Ca + Calcitonin
0.04 (−0.04, 0.12)
Very low
Percent BMD change at the lumbar spine (466)
 Bis+Ca vs.  Bis+Ca + Vit D
−3.27 (−7.87, 0.84)
Very low
 Bis+Ca vs. Ca
4 (152)
1.17 (0.80, 1.54)
7.24 (3.73, 10.69)
91.70%
0.000
Moderate
 Bis+Ca vs. Ca + Vit D
2 (46)
1.53 (0.79, 2.27)
2.22 (−1.44, 5.73)
94.10%
0.000
Low
 Bis+Ca vs. Ca + Calcitonin
1 (31)
3.13 (−2.51, 8.51)
Low
 Bis+Ca + Vit D vs. Ca
10.50 (5.92, 15.34)
Very low
 Bis+Ca + Vit D vs. Ca + Vit D
3 (145)
1.32 (1.02, 1.62)
5.48 (2.57, 8.42)
98.30%
0.000
Moderate
 Bis+Ca + Vit D vs. Ca + Calcitonin
6.39 (0.55, 12.89)
Low
 Ca vs. Ca + Vit D
2 (51)
−2.73 (−3.51, −1.95)
−5.02 (−8.84, − 1.20)
0.00%
0.373
Moderate
 Ca vs. Ca + Calcitonin
1 (30)
−4.11 (−9.01, 0.72)
Low
 Ca + Vit D vs. Ca + Calcitonin
0.91 (−4.38, 6.44)
Very low
Bis = bisphosphonate, Ca = calcium, Vit D = Vitamin D analogs, 95% CI = 95% Confidence Intervals, 95% CrI = 95% Credible Intervals. The mean difference with 95% CI or 95% CrI was used for continuous outcomes. Significant results are in bold. The Grading of Recommendations Assessment, Development and Evaluation (GRADE) approach specific to NMA served to assess the certainty in the evidence (quality of evidence) associated with specific comparisons, including direct, indirect, and final network meta-analysis estimates. The confidence assessment addressed the risk of bias (in individual studies), imprecision, inconsistency (heterogeneity in estimates of effect across studies), indirectness, and publication bias

Network meta-analysis— Primary outcome

Change of BMD at the femoral neck

Ten RCTs involving 536 adults evaluated the absolute change in BMD at the femoral neck. Figure 2 summarizes the network of direct evidence available for this outcome. No statistically significant difference was detected between each treatment groups. The SUCRA value for the regimens were 88%, 53%, 52%, 29%, 28% for bisphosphonate with calcium, bisphosphonate with calcium and vitamin D analogs, calcium with vitamin D analogs, calcitonin with calcium and calcium (Fig. 3a).
The result of percent terms was reported by 5 RCTs including 284 patients. Only bisphosphonate plus calcium revealed a significant gain in percent BMD change than calcium alone (MD, 5.83; 95% CrI, 1.61 to 9.27). No statistical difference was observed between other groups. Bisphosphonate combined with calcium and vitamin D analogs had the highest SUCRA value (97% Fig. 3b), followed by calcitonin with calcium (66%), bisphosphonate plus calcium (38%), calcium with vitamin D analogs (27%), and calcium only (22%).

Change of BMD at the lumbar spine

14 RCTs including 814 participants provided data for comparison of absolute change in BMD at the lumbar spine. Bisphosphonate and calcium with or without vitamin D analogs outperformed calcium solely (MD, 0.07; 95% CrI, 0.00 to 0.13; MD, 0.06; 95% CrI, 0.02 to 0.09). We also observed that compared to calcium with vitamin D analogs, adding bisphosphonate was associated with marked improvement (MD, 0.03; 95% CrI, 0.00 to 0.05). The SUCRA value for each treatment formulations were as follows (Fig. 3c): bisphosphonate with calcium and vitamin D analogs (87%), bisphosphonate with calcium (81%), calcium plus vitamin D analogs (48%), calcitonin with calcium (24%) and calcium solely (10%).
Considering percent terms, the result analyzed using data from 7 trials (466 patients). Combination of bisphosphonate with calcium and vitamin D analogs showed greater beneficial effects than calcium alone or with either vitamin D analogs or calcitonin (MD, 10.51; 95% CrI, 5.92 to 15.34; MD, 5.48; 95% CrI, 2.57 to 8.42; MD, 6.39; 95% CrI, 0.55 to 12.89). Both bisphosphonate and vitamin D analogs combined with calcium displayed a notable improvement compared to calcium alone (MD, 7.24; 95% CrI, 3.73 to 10.69; MD, 5.02; 95% CrI, 1.20 to 8.84). As expected, bisphosphonate combined with calcium and vitamin D analogs had the highest SUCRA value (Fig. 3d 99%), followed by bisphosphonate with calcium (63%), calcium with vitamin D analogs (57%), calcitonin with calcium (29%), and calcium only (2%).

Secondary outcomes

We did not observe a significant difference in the incidence of fractures from the direct comparisons and it could not connect to draw network geometries. All treatments have uncertain effects on all-cause mortality and graft loss metrics. Similarly, there were no statistical differences in the number of biopsy-proven acute rejections as well as adverse events among treatment groups. However, we found more adverse events happened in bisphosphonate and calcium than in calcium alone (OR, 5.41; 95% CrI, 1.15 to 25.49) from pairwise meta-analysis. Further details of the secondary outcome analyses are presented in Additional files 3 and 4.

Network consistency

No evidence of small study effects based on funnel plot asymmetry was observed, but the number of studies included in each comparison was small. There was no evidence of inconsistency in the NMA when we applied the node-splitting approach. The total residual deviance for the outcomes of percent change (23.73, pD = 22) and absolute change (43.86, pD = 45) of BMD at the lumbar spine implied a good model fit, as well as percent change (25.22, pD = 26) and absolute change (24.36, pD = 24) at the femoral neck.

Sensitivity analysis

For the sensitivity analyses, we used the full network for the primary outcome. In the first analysis, we investigated the different assumptions regarding the potential relationship between time and treatment effect, Bayesian NMA were repeated using the absolute change of BMD at the twelve-month follow-up period. We observed comparable results at the lumbar spine, adding bisphosphonate showed significant improvement than calcium alone or calcium with vitamin D analogs (Additional file 5: MD, 0.06; 95% CrI, 0.01 to 0.10; MD, 0.03; 95% CrI, 0.00 to 0.07). We also observed that bisphosphonate with calcium and vitamin D analogs outperformed calcium solely (MD, 0.07; 95% CrI, 0.01 to 0.15). At the femoral neck, bisphosphonate with calcium showed a significant preference than calcium alone (Additional file 5; MD, 0.23; 95% CrI, 0.02 to 0.46). The parameter estimates were consistent with the main analysis. We carried out separate meta-regressions to test the effect of length of trial, publication date and modes of administration. No evidence exists for an interaction between any of the trial characteristics assessed and the treatment effect.

Quality of evidence

In general, there was no serious risk of bias, indirectness, inconsistency, or publication bias for any of the direct comparisons. In several comparisons, there was serious imprecision in summary estimate because the 95% credible interval crossed unity. The GRADE quality of evidence supporting the use of each treatment for the primary outcome was outlined in Table 2. According to GRADE, we had moderate confidence in estimates supporting the combination use of bisphosphonate or vitamin D analogs with calcium for improving BMD at the lumbar spine. We detected using bisphosphonate combined with vitamin D analogs and calcium considering BMD change at the lumbar spine with low quality evidence. There was very confidence in estimates supporting using calcitonin with calcium both at the lumbar spine and the femoral neck. Conceptually, there was no significant intransitivity.

Discussion

This NMA was aimed to investigate the comparable efficacy and safety of bisphosphonate and its co-interventions for the post-transplantation bone disease. We found the combination of bisphosphonate, calcium and vitamin D analogs was the most effective to prevent bone and restore or improve BMD. However, the effects on fracture risk, adverse events, death, acute renal rejection and graft loss were still uncertain because of insufficient data and short follow-up time.
The current study revealed that only calcium prescription could not benefit KTRs from bone loss. We may suggest KTRs take calcium and vitamin D analogs orally because this NMA showed combination therapy of calcium and vitamin D analogs could improve BMD than calcium alone with moderate quality evidence. The result was supported by previous studies [37, 38]. Our work displayed that calcitonin with calcium seemed only better than calcium alone with low strength of evidence. Two RCTs [26, 33] involving 31 patients allocated to receive calcitonin, and incidence of hypocalcemia was reported so that we did not suggest giving calcitonin for KTRs. In our study, additional use of bisphosphonate could improve BMD changes at the lumbar spine and femoral neck. It was in accordance with previous analyses [9, 39], though they did not examine the effect of calcium and vitamin D analogs supplementation. Importantly, the validity and robustness of NMA depends not only on the heterogeneity in case of standard pairwise meta-analysis, but also on the inconsistency between direct and indirect contrast estimates. No evidence of inconsistency was found in this NMA. Bisphosphonate plus calcium revealed a significant gain in percent BMD change than calcium alone. The heterogeneity was calculated from the pairwise meta-analysis with four RCTs [3133, 35]. The sample size, races and bisphosphonates which included ibandronate [31], pamidronate [32], clodronate [33], alendronate [35] were different. These resulted in high heterogeneity which would reduce the quality of evidence. Adding bisphosphonates also showed significant improvement than combination of calcium and vitamin D analogs in both absolute and percent BMD change at the lumbar with high heterogeneity. The heterogeneity was associated with characteristics of samples, different inclusion and exclusion criterion. The quality of evidence downgraded because of high heterogeneity. At this stage, limited information made it difficult to perform further analysis. Thus, we could not ignore the impact of heterogeneity when draw the conclusion. Although bisphosphonate with calcium and vitamin D analogs ranked the best, we did not detect any significant differences between combination use of bisphosphonate and calcium with or without vitamin D analogs from the indirect comparisons. Moreover, indirect comparisons would lead to very low quality of evidence. Only RCT conducted by Fan SL et al. [40, 41] compared bisphosphonate alone with placebo. They found that only two intravenous doses of pamidronate can protect the skeleton from bone loss even 4 years later after transplantation. We could not specify the influence of bisphosphonate monotherapy and access the situation of KTRs with no treatment due to lack of relevant studies.
Included RCTs used BMD as a surrogate marker and did not provide sufficient data to make a polygonal network configuration about the fracture. Also, the association between BMD metrics and fracture risk in KTRs is still controversial. West SL et al. [42] indicated that low BMD was a risk factor for subsequent fracture in patients with pre-dialysis CKD, but data for KTRs are scant. According to KDIGO guideline [7], bone biopsy is reasonable to guide treatment in the first twelve-months after transplantation. However, it is an invasive procedure and most centers lack the expertise to properly process and analyze bone biopsy specimens. Recently, the Fracture Risk Assessment Tool (FRAX) [43] and the spine Trabecular bone score (TBS) [44] were detected as new measurements for KTRs to predict fracture risk. Consequently, surrogate outcomes poorly reflect pathological bone changes. Future trials need to find more specific measurements for detecting mineral and bone disorders in KTRs.
Evidence on other secondary outcomes was limited. There was an unexpected finding from previous reviews [9, 45] that bisphosphonate reduced acute graft rejection moderately. Bisphosphonates could suppress cytokine releasing from activated macrophages to inhibit T-cell function. Its immunomodulatory and anti-inflammatory properties may explain this finding. However, the confidence intervals were wide and ignored the influence of co-interventions. The use of bisphosphonate was limited on account of its nephrotoxicity and development of the adynamic bone disease. We did not find additional bisphosphonate use would increase the occurrence of graft loss and adverse events. Apart from mild gastrointestinal side effects, include RCTs did not report or systematically study serious adverse events. Perazella MA et al. [46] summarized that bisphosphonate nephrotoxicity is infusion time-dependent and dose-dependent. Increasing the time interval between doses can limit its nephrotoxicity. On the current situation, bisphosphonate therapy was well tolerated whereas controversial data on its potency in preventing fracture limited its widespread.
Our analysis updated the previous meta-analysis and conducted a comprehensive search with broad inclusion criteria to maximize available data in this field. Only RCTs that supplied BMD results with g/cm2 were included to standardize each comparison, while some studies used different units such as Z-score or T-score. Furthermore, we used GRADE approach to measure the quality of evidence and also performed sensitivity analyses to demonstrate the robustness of estimates. In addition, only adult KTRs were included to offer more reliable evidence and minimize potential bias. To our knowledge, this is the first NMA that took co-intervention (calcium, vitamin D analogs) into account when examining the effect of bisphosphonates and expands on previous meta-analyses as well [9, 45].
However, this NMA still has several limitations including the omission of important methodological details in RCTs and the possibility of reporting biases. Most included studies had a high risk of bias and their impact on results is uncertain. Moreover, some studies only included cadaveric allograft, while some studies excluded patients with diabetes or postmenopausal women. These were risk factors for fracture. Preexisting CKD-MBD, immunosuppression therapy including steroid dosage, CNIs type could also cause bone disease after KT. Because of insufficient information, we could not perform further analysis to identify the influence of relevant factors. These would result in high heterogeneity which may downgrade the quality of evidence as well. Aside from different basic characteristic of the participant, it is unknown if within the drug class of bisphosphonates certain drugs are more favorable than others, and the bisphosphonates regimen (dosage, route, timing, and administration duration) differ among the included studies. We grouped vitamin D, calcitriol, alfacalcidol together as vitamin D analogs and did not distinguish their efficacy. These factors may potentially influence the calculation of BMD between RCTs.
More high-quality RCT is required to determine the optimal therapy for KTRs to prevent fractures with minimal risk for side effects. We also need to find a more correlative measurement than BMD to reflect pathological bone changes in KTRs. Future studies should be powered to show the fracture risk with sufficient follow-up time (≥3 years) and adequate sample sizes, while providing methodological details.

Conclusion

At this stage, we suggested the additional use of bisphosphonate was well-tolerated and more favorable in KTRs to improve BMD at the lumbar spine and femoral neck. However, evidence to reduce fracture risk is insufficient. Clinicians should take all known safety information and compliance of patients into account when using bisphosphonates. Further studies are needed to support our findings and find optimal treatment option for KTRs.

Acknowledgments

The authors would like to acknowledge Ian Charles Tobias for reviewing the manuscript.

Funding

This study was supported by the National Natural Science Foundation of China (Grant No.81570668).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
All analyses were based on previous published studies, thus no ethical approval and patient consent are required.
Not applicable.

Competing interests

The author declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Mitterbauer C, Oberbauer R. Bone disease after kidney transplantation. Transpl Int. 2008;21(7):615–24.CrossRef Mitterbauer C, Oberbauer R. Bone disease after kidney transplantation. Transpl Int. 2008;21(7):615–24.CrossRef
2.
Zurück zum Zitat Naylor KL, Li AH, Lam NN, Hodsman AB, Jamal SA, Garg AX. Fracture risk in kidney transplant recipients: a systematic review. Transplantation. 2013;95(12):1461–70.CrossRef Naylor KL, Li AH, Lam NN, Hodsman AB, Jamal SA, Garg AX. Fracture risk in kidney transplant recipients: a systematic review. Transplantation. 2013;95(12):1461–70.CrossRef
3.
Zurück zum Zitat Weisinger JR, Carlini RG, Rojas E, Bellorin-Font E. Bone disease after renal transplantation. Clin J Am Soc Nephrol. 2006;1(6):1300–13.CrossRef Weisinger JR, Carlini RG, Rojas E, Bellorin-Font E. Bone disease after renal transplantation. Clin J Am Soc Nephrol. 2006;1(6):1300–13.CrossRef
4.
Zurück zum Zitat Bouquegneau A, Salam S, Delanaye P, Eastell R, Khwaja A. Bone disease after kidney transplantation. Clin J Am Soc Nephrol. 2016;11(7):1282–96.CrossRef Bouquegneau A, Salam S, Delanaye P, Eastell R, Khwaja A. Bone disease after kidney transplantation. Clin J Am Soc Nephrol. 2016;11(7):1282–96.CrossRef
5.
Zurück zum Zitat Cunningham J. Pathogenesis and prevention of bone loss in patients who have kidney disease and receive long-term immunosuppression. J Am Soc Nephrol. 2007;18(1):223–34.CrossRef Cunningham J. Pathogenesis and prevention of bone loss in patients who have kidney disease and receive long-term immunosuppression. J Am Soc Nephrol. 2007;18(1):223–34.CrossRef
6.
Zurück zum Zitat Evenepoel P, Cooper K, Holdaas H, Messa P, Mourad G, Olgaard K, Rutkowski B, Schaefer H, Deng H, Torregrosa JV, Wuthrich RP, Yue S. A randomized study evaluating cinacalcet to treat hypercalcemia in renal transplant recipients with persistent hyperparathyroidism. Am J Transplant. 2014;14(11):2545–55.CrossRef Evenepoel P, Cooper K, Holdaas H, Messa P, Mourad G, Olgaard K, Rutkowski B, Schaefer H, Deng H, Torregrosa JV, Wuthrich RP, Yue S. A randomized study evaluating cinacalcet to treat hypercalcemia in renal transplant recipients with persistent hyperparathyroidism. Am J Transplant. 2014;14(11):2545–55.CrossRef
7.
Zurück zum Zitat Ketteler M, Block GA, Evenepoel P, Fukagawa M, Herzog CA, McCann L, Moe SM, Shroff R, Tonelli MA, Toussaint ND, Vervloet MG, Leonard MB. Executive summary of the 2017 KDIGO chronic kidney disease-mineral and bone disorder (CKD-MBD) guideline update: what's changed and why it matters. Kidney Int. 2017;92(1):26–36.CrossRef Ketteler M, Block GA, Evenepoel P, Fukagawa M, Herzog CA, McCann L, Moe SM, Shroff R, Tonelli MA, Toussaint ND, Vervloet MG, Leonard MB. Executive summary of the 2017 KDIGO chronic kidney disease-mineral and bone disorder (CKD-MBD) guideline update: what's changed and why it matters. Kidney Int. 2017;92(1):26–36.CrossRef
8.
Zurück zum Zitat Palmer SC, McGregor DO, Strippoli GF. Interventions for preventing bone disease in kidney transplant recipients. Cochrane Database Syst Rev. 2007. Palmer SC, McGregor DO, Strippoli GF. Interventions for preventing bone disease in kidney transplant recipients. Cochrane Database Syst Rev. 2007.
9.
Zurück zum Zitat Versele EB, Van Laecke S, Dhondt AW, Verbeke F, Vanholder R, Van Biesen W, Nagler EV. Bisphosphonates for preventing bone disease in kidney transplant recipients: a meta-analysis of randomized controlled trials. Transpl Int. 2015;29(2):153–64.CrossRef Versele EB, Van Laecke S, Dhondt AW, Verbeke F, Vanholder R, Van Biesen W, Nagler EV. Bisphosphonates for preventing bone disease in kidney transplant recipients: a meta-analysis of randomized controlled trials. Transpl Int. 2015;29(2):153–64.CrossRef
10.
Zurück zum Zitat Hutton B, Salanti G, Caldwell DM, Chaimani A, Schmid CH, Cameron C, Ioannidis JP, Straus S, Thorlund K, Jansen JP, Mulrow C, Catalá-López F, Gøtzsche PC, Dickersin K, Boutron I, Altman DG, Moher D. The PRISMA extension statement for reporting of systematic reviews incorporating network meta-analyses of health care interventions: checklist and explanations. Ann Intern Med. 2015;162(11):777–84.CrossRef Hutton B, Salanti G, Caldwell DM, Chaimani A, Schmid CH, Cameron C, Ioannidis JP, Straus S, Thorlund K, Jansen JP, Mulrow C, Catalá-López F, Gøtzsche PC, Dickersin K, Boutron I, Altman DG, Moher D. The PRISMA extension statement for reporting of systematic reviews incorporating network meta-analyses of health care interventions: checklist and explanations. Ann Intern Med. 2015;162(11):777–84.CrossRef
11.
Zurück zum Zitat Higgins G, Green S. Cochrane handbook for systematic reviews of interventions version 5.1.0. Updated march, 2011. London: The Cochrane Collaboration; 2011. Higgins G, Green S. Cochrane handbook for systematic reviews of interventions version 5.1.0. Updated march, 2011. London: The Cochrane Collaboration; 2011.
12.
Zurück zum Zitat DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. 1986;7:177–88.CrossRef DerSimonian R, Laird N. Meta-analysis in clinical trials. Control Clin Trials. 1986;7:177–88.CrossRef
13.
Zurück zum Zitat Salanti G, Higgins JP, Ades AE, Ioannidis JP. Evaluation of networks of randomised trials. Stat Methods Med Res. 2008;17(3):279–301.CrossRef Salanti G, Higgins JP, Ades AE, Ioannidis JP. Evaluation of networks of randomised trials. Stat Methods Med Res. 2008;17(3):279–301.CrossRef
14.
Zurück zum Zitat Higgins JP, Jackson D, Barrett JK, Lu G, Ades AE, White IR. Consistency and inconsistency in network meta-analysis: concepts and models for multi-arm studies. Res Synth Methods. 2012;3(2):98–110.CrossRef Higgins JP, Jackson D, Barrett JK, Lu G, Ades AE, White IR. Consistency and inconsistency in network meta-analysis: concepts and models for multi-arm studies. Res Synth Methods. 2012;3(2):98–110.CrossRef
15.
Zurück zum Zitat Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ. 2003;327(7414):557–60.CrossRef Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ. 2003;327(7414):557–60.CrossRef
16.
Zurück zum Zitat Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, Schünemann HJ, GRADE Working Group. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ. 2008;336(7650):924–6.CrossRef Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, Schünemann HJ, GRADE Working Group. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ. 2008;336(7650):924–6.CrossRef
17.
Zurück zum Zitat Salanti G, Del Giovane C, Chaimani A, Caldwell DM, Higgins JP. Evaluating the quality of evidence from a network meta-analysis. PLoS One. 2014;9(7):e99682.CrossRef Salanti G, Del Giovane C, Chaimani A, Caldwell DM, Higgins JP. Evaluating the quality of evidence from a network meta-analysis. PLoS One. 2014;9(7):e99682.CrossRef
18.
Zurück zum Zitat Smerud KT, Dolgos S, Olsen IC, Åsberg A, Sagedal S, Reisæter AV, Midtvedt K, Pfeffer P, Ueland T, Godang K, Bollerslev J, Hartmann A. A 1-year randomized, double-blind, placebo-controlled study of intravenous ibandronate on bone loss following renal transplantation. Am J Transplant. 2012;12(12):3316–25.CrossRef Smerud KT, Dolgos S, Olsen IC, Åsberg A, Sagedal S, Reisæter AV, Midtvedt K, Pfeffer P, Ueland T, Godang K, Bollerslev J, Hartmann A. A 1-year randomized, double-blind, placebo-controlled study of intravenous ibandronate on bone loss following renal transplantation. Am J Transplant. 2012;12(12):3316–25.CrossRef
19.
Zurück zum Zitat Coco M, Pullman J, Cohen HW, Lee S, Shapiro C, Solorzano C, Greenstein S, Glicklich D. Effect of risedronate on bone in renal transplant recipients. J Am Soc Nephrol. 2012;23(8):1426–37.CrossRef Coco M, Pullman J, Cohen HW, Lee S, Shapiro C, Solorzano C, Greenstein S, Glicklich D. Effect of risedronate on bone in renal transplant recipients. J Am Soc Nephrol. 2012;23(8):1426–37.CrossRef
20.
Zurück zum Zitat Torregrosa JV, Fuster D, Gentil MA, Marcen R, Guirado L, Zarraga S, Bravo J, Burgos D, Monegal A, Muxí A, García S. Open-label trial: effect of weekly risedronate immediately after transplantation in kidney recipients. Transplantation. 2010;89(12):1476–81.CrossRef Torregrosa JV, Fuster D, Gentil MA, Marcen R, Guirado L, Zarraga S, Bravo J, Burgos D, Monegal A, Muxí A, García S. Open-label trial: effect of weekly risedronate immediately after transplantation in kidney recipients. Transplantation. 2010;89(12):1476–81.CrossRef
21.
Zurück zum Zitat Torregrosa JV, Fuster D, Monegal A, Gentil MA, Bravo J, Guirado L, Muxí A, Cubero J. Efficacy of low doses of pamidronate in osteopenic patients administered in the early post-renal transplant. Osteoporos Int. 2011;22(1):281–7.CrossRef Torregrosa JV, Fuster D, Monegal A, Gentil MA, Bravo J, Guirado L, Muxí A, Cubero J. Efficacy of low doses of pamidronate in osteopenic patients administered in the early post-renal transplant. Osteoporos Int. 2011;22(1):281–7.CrossRef
22.
Zurück zum Zitat Walsh SB, Altmann P, Pattison J, Wilkie M, Yaqoob MM, Dudley C, Cockwell P, Sweny P, Banks LM, Hall-Craggs M, Noonan K, Andrews C, Cunningham J. Effect of pamidronate on bone loss after kidney transplantation: a randomized trial. Am J Kidney Dis. 2009;53(5):856–65.CrossRef Walsh SB, Altmann P, Pattison J, Wilkie M, Yaqoob MM, Dudley C, Cockwell P, Sweny P, Banks LM, Hall-Craggs M, Noonan K, Andrews C, Cunningham J. Effect of pamidronate on bone loss after kidney transplantation: a randomized trial. Am J Kidney Dis. 2009;53(5):856–65.CrossRef
23.
Zurück zum Zitat Lan G, Peng L, Xie X, Peng F, Wang Y, Yu S. Alendronate is effective to treat bone loss in renal transplantation recipients. Transplant Proc. 2008;40(10):3496–8.CrossRef Lan G, Peng L, Xie X, Peng F, Wang Y, Yu S. Alendronate is effective to treat bone loss in renal transplantation recipients. Transplant Proc. 2008;40(10):3496–8.CrossRef
24.
Zurück zum Zitat Trabulus S, Altiparmak MR, Apaydin S, Serdengecti K, Sariyar M. Treatment of renal transplant recipients with low bone mineral density: a randomized prospective trial of alendronate, alfacalcidol, and alendronate combined with alfacalcidol. Transplant Proc. 2008;40(1):160–6.CrossRef Trabulus S, Altiparmak MR, Apaydin S, Serdengecti K, Sariyar M. Treatment of renal transplant recipients with low bone mineral density: a randomized prospective trial of alendronate, alfacalcidol, and alendronate combined with alfacalcidol. Transplant Proc. 2008;40(1):160–6.CrossRef
25.
Zurück zum Zitat Nayak B, Guleria S, Varma M, Tandon N, Aggarwal S, Bhowmick D, Agarwal SK, Mahajan S, Gupta S, Tiwari SC. Effect of bisphosphonates on bone mineral density after renal transplantation as assessed by bone mineral densitometry. Transplant Proc. 2007;39(3):750–2.CrossRef Nayak B, Guleria S, Varma M, Tandon N, Aggarwal S, Bhowmick D, Agarwal SK, Mahajan S, Gupta S, Tiwari SC. Effect of bisphosphonates on bone mineral density after renal transplantation as assessed by bone mineral densitometry. Transplant Proc. 2007;39(3):750–2.CrossRef
26.
Zurück zum Zitat El-Agroudy AE, El-Husseini AA, El-Sayed M, Mohsen T, Ghoneim MA. A prospective randomized study for prevention of postrenal transplantation bone loss. Kidney Int. 2005;67(5):2039–45.CrossRef El-Agroudy AE, El-Husseini AA, El-Sayed M, Mohsen T, Ghoneim MA. A prospective randomized study for prevention of postrenal transplantation bone loss. Kidney Int. 2005;67(5):2039–45.CrossRef
27.
Zurück zum Zitat Schwarz C, Mitterbauer C, Heinze G, Woloszczuk W, Haas M, Oberbauer R. Nonsustained effect of short-term bisphosphonate therapy on bone turnover three years after renal transplantation. Kidney Int. 2004;65(1):304–9.CrossRef Schwarz C, Mitterbauer C, Heinze G, Woloszczuk W, Haas M, Oberbauer R. Nonsustained effect of short-term bisphosphonate therapy on bone turnover three years after renal transplantation. Kidney Int. 2004;65(1):304–9.CrossRef
28.
Zurück zum Zitat Jeffery JR, Leslie WD, Karpinski ME, Nickerson PW, Rush DN. Prevalence and treatment of decreased bone density in renal transplant recipients: a randomized prospective trial of calcitriol versus alendronate. Transplantation. 2003;76(10):1498–502.CrossRef Jeffery JR, Leslie WD, Karpinski ME, Nickerson PW, Rush DN. Prevalence and treatment of decreased bone density in renal transplant recipients: a randomized prospective trial of calcitriol versus alendronate. Transplantation. 2003;76(10):1498–502.CrossRef
29.
Zurück zum Zitat Coco M, Glicklich D, Faugere MC, Burris L, Bognar I, Durkin P, Tellis V, Greenstein S, Schechner R, Figueroa K, McDonough P, Wang G, Malluche H. Prevention of bone loss in renal transplant recipients: a prospective, randomized trial of intravenous pamidronate. J Am Soc Nephrol. 2003;14(10):2669–76.CrossRef Coco M, Glicklich D, Faugere MC, Burris L, Bognar I, Durkin P, Tellis V, Greenstein S, Schechner R, Figueroa K, McDonough P, Wang G, Malluche H. Prevention of bone loss in renal transplant recipients: a prospective, randomized trial of intravenous pamidronate. J Am Soc Nephrol. 2003;14(10):2669–76.CrossRef
30.
Zurück zum Zitat Haas M, Leko-Mohr Z, Roschger P, Kletzmayr J, Schwarz C, Mitterbauer C, Steininger R, Grampp S, Klaushofer K, Delling G, Oberbauer R. Zoledronic acid to prevent bone loss in the first 6 months after renal transplantation. Kidney Int. 2003;63(3):1130–6.CrossRef Haas M, Leko-Mohr Z, Roschger P, Kletzmayr J, Schwarz C, Mitterbauer C, Steininger R, Grampp S, Klaushofer K, Delling G, Oberbauer R. Zoledronic acid to prevent bone loss in the first 6 months after renal transplantation. Kidney Int. 2003;63(3):1130–6.CrossRef
31.
Zurück zum Zitat Grotz W, Nagel C, Poeschel D, Cybulla M, Petersen KG, Uhl M, Strey C, Kirste G, Olschewski M, Reichelt A, Rump LC. Effect of Ibandronate on bone loss and renal function after kidney transplantation. J Am Soc Nephrol. 2001;12(7):1530–7.PubMed Grotz W, Nagel C, Poeschel D, Cybulla M, Petersen KG, Uhl M, Strey C, Kirste G, Olschewski M, Reichelt A, Rump LC. Effect of Ibandronate on bone loss and renal function after kidney transplantation. J Am Soc Nephrol. 2001;12(7):1530–7.PubMed
32.
Zurück zum Zitat Nam JH, Moon JI, Chung SS, Kim SI, Park KI, Song YD, Kim KR, Lee HC, Huh K, Lim SK. Pamidronate and calcitriol trial for the prevention of early bone loss after renal transplantation. Transplant Proc. 2000;32(7):1876.CrossRef Nam JH, Moon JI, Chung SS, Kim SI, Park KI, Song YD, Kim KR, Lee HC, Huh K, Lim SK. Pamidronate and calcitriol trial for the prevention of early bone loss after renal transplantation. Transplant Proc. 2000;32(7):1876.CrossRef
33.
Zurück zum Zitat Grotz WH, Rump LC, Niessen A, Schmidt-Gayk H, Reichelt A, Kirste G, Olschewski M, Schollmeyer PJ. Treatment of osteopenia and osteoporosis after kidney transplantation. Transplantation. 1998;66(8):1004–8.CrossRef Grotz WH, Rump LC, Niessen A, Schmidt-Gayk H, Reichelt A, Kirste G, Olschewski M, Schollmeyer PJ. Treatment of osteopenia and osteoporosis after kidney transplantation. Transplantation. 1998;66(8):1004–8.CrossRef
34.
Zurück zum Zitat Giannini S, D'Angelo A, Carraro G, Nobile M, Rigotti P, Bonfante L, Marchini F, Zaninotto M, Dalle Carbonare L, Sartori L, Crepaldi G. Alendronate prevents further bone loss in renal transplant recipients. J Bone Miner Res. 2001;16(11):2111–7.CrossRef Giannini S, D'Angelo A, Carraro G, Nobile M, Rigotti P, Bonfante L, Marchini F, Zaninotto M, Dalle Carbonare L, Sartori L, Crepaldi G. Alendronate prevents further bone loss in renal transplant recipients. J Bone Miner Res. 2001;16(11):2111–7.CrossRef
35.
Zurück zum Zitat Koc M, Tuglular S, Arikan H, Ozener C, Akoglu E. Alendronate increases bone mineral density in long-term renal transplant recipients. Transplant Proc. 2002;34(6):2111–3.CrossRef Koc M, Tuglular S, Arikan H, Ozener C, Akoglu E. Alendronate increases bone mineral density in long-term renal transplant recipients. Transplant Proc. 2002;34(6):2111–3.CrossRef
36.
Zurück zum Zitat Torregrosa JV, Fuster D, Pedroso S, Diekmann F, Campistol JM, Rubí S, Oppenheimer F. Weekly risedronate in kidney transplant patients with osteopenia. Transpl Int. 2007;20(8):708–11.CrossRef Torregrosa JV, Fuster D, Pedroso S, Diekmann F, Campistol JM, Rubí S, Oppenheimer F. Weekly risedronate in kidney transplant patients with osteopenia. Transpl Int. 2007;20(8):708–11.CrossRef
37.
Zurück zum Zitat Elagroudy AE, et al. Preventing bone loss in renal transplant recipients with vitamin D. J Am Soc Nephrol. 2003;14(11):2975–9.CrossRef Elagroudy AE, et al. Preventing bone loss in renal transplant recipients with vitamin D. J Am Soc Nephrol. 2003;14(11):2975–9.CrossRef
38.
Zurück zum Zitat Josephson MA, Schumm LP, Chiu MY, Marshall C, Thistlethwaite JR, Sprague SM. Calcium and calcitriol prophylaxis attenuates posttransplant bone loss. Transplantation. 2004;78(8):1233–6.CrossRef Josephson MA, Schumm LP, Chiu MY, Marshall C, Thistlethwaite JR, Sprague SM. Calcium and calcitriol prophylaxis attenuates posttransplant bone loss. Transplantation. 2004;78(8):1233–6.CrossRef
39.
Zurück zum Zitat Kan SL, Ning GZ, Chen LX, Zhou Y, Sun JC, Feng SQ. Efficacy and safety of bisphosphonates for low bone mineral density after kidney transplantation: a meta-analysis. Medicine. 2016;95(5):e2679.CrossRef Kan SL, Ning GZ, Chen LX, Zhou Y, Sun JC, Feng SQ. Efficacy and safety of bisphosphonates for low bone mineral density after kidney transplantation: a meta-analysis. Medicine. 2016;95(5):e2679.CrossRef
40.
Zurück zum Zitat Fan SL, Almond MK, Ball E, Evans K, Cunningham J. Pamidronate therapy as prevention of bone loss following renal transplantation. Kidney Int. 2000;57(2):684–90.CrossRef Fan SL, Almond MK, Ball E, Evans K, Cunningham J. Pamidronate therapy as prevention of bone loss following renal transplantation. Kidney Int. 2000;57(2):684–90.CrossRef
41.
Zurück zum Zitat Fan SL, Kumar S, Cunningham J. Long-term effects on bone mineral density of pamidronate given at the time of renal transplantation. Kidney Int. 2003;63(6):2275–9.CrossRef Fan SL, Kumar S, Cunningham J. Long-term effects on bone mineral density of pamidronate given at the time of renal transplantation. Kidney Int. 2003;63(6):2275–9.CrossRef
42.
Zurück zum Zitat West SL, Lok CE, Langsetmo L, Cheung AM, Szabo E, Pearce D, Fusaro M, Wald R, Weinstein J, Jamal SA. Bone mineral density predicts fractures in chronic kidney disease. J Bone Miner Res. 2015;30(5):913–9.CrossRef West SL, Lok CE, Langsetmo L, Cheung AM, Szabo E, Pearce D, Fusaro M, Wald R, Weinstein J, Jamal SA. Bone mineral density predicts fractures in chronic kidney disease. J Bone Miner Res. 2015;30(5):913–9.CrossRef
43.
Zurück zum Zitat Naylor KL, Leslie WD, Hodsman AB, Rush DN, Garg AX. FRAX predicts fracture risk in kidney transplant recipients. Transplantation. 2014;97(9):940–5.CrossRef Naylor KL, Leslie WD, Hodsman AB, Rush DN, Garg AX. FRAX predicts fracture risk in kidney transplant recipients. Transplantation. 2014;97(9):940–5.CrossRef
44.
Zurück zum Zitat Luckman M, Hans D, Cortez N, Nishiyama KK, Agarawal S, Zhang C, Nikkel L, Iyer S, Fusaro M, Guo EX, McMahon DJ, Shane E, Nickolas TL. Spine trabecular bone score as an Indicator of bone microarchitecture at the peripheral skeleton in kidney transplant recipients. Clin J Am Soc Nephrol. 2017;12(4):644–52.CrossRef Luckman M, Hans D, Cortez N, Nishiyama KK, Agarawal S, Zhang C, Nikkel L, Iyer S, Fusaro M, Guo EX, McMahon DJ, Shane E, Nickolas TL. Spine trabecular bone score as an Indicator of bone microarchitecture at the peripheral skeleton in kidney transplant recipients. Clin J Am Soc Nephrol. 2017;12(4):644–52.CrossRef
45.
Zurück zum Zitat Palmer SC, Mcgregor DO, Strippoli GF. Interventions for preventing bone disease in kidney transplant recipients: a systematic review of randomized controlled trials. Am J Kidney Dis. 2005;45(4):638–49.CrossRef Palmer SC, Mcgregor DO, Strippoli GF. Interventions for preventing bone disease in kidney transplant recipients: a systematic review of randomized controlled trials. Am J Kidney Dis. 2005;45(4):638–49.CrossRef
46.
Zurück zum Zitat Perazella MA, Markowitz GS. Bisphosphonate nephrotoxicity. Kidney Int. 2008;74(11):1385–93.CrossRef Perazella MA, Markowitz GS. Bisphosphonate nephrotoxicity. Kidney Int. 2008;74(11):1385–93.CrossRef
Metadaten
Titel
Outcomes of bisphosphonate and its supplements for bone loss in kidney transplant recipients: a systematic review and network meta-analysis
verfasst von
Yan Yang
Shi Qiu
Linghui Deng
Xi Tang
Xinrui Li
Qiang Wei
Ping Fu
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Nephrology / Ausgabe 1/2018
Elektronische ISSN: 1471-2369
DOI
https://doi.org/10.1186/s12882-018-1076-1

Weitere Artikel der Ausgabe 1/2018

BMC Nephrology 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

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