Skip to main content
Erschienen in: BMC Nephrology 1/2022

Open Access 01.12.2022 | Research

Recipient obesity as a risk factor in kidney transplantation

verfasst von: Uwe Scheuermann, Jonas Babel, Uta-Carolin Pietsch, Antje Weimann, Orestis Lyros, Katrin Semmling, Hans-Michael Hau, Daniel Seehofer, Sebastian Rademacher, Robert Sucher

Erschienen in: BMC Nephrology | Ausgabe 1/2022

Abstract

Background

The aim of the study was to investigate the effect of recipient obesity on the short- and long-term outcomes of patients undergoing primary kidney transplantation (KT).

Patients and methods

A total of 578 patients receiving primary KT in our department between 1993 and 2017 were included in the study. Patients were divided according to their body mass index (BMI) into normal weight (BMI 18.5–24.9 kg/m2; N = 304), overweight (BMI 25–29.9 kg/m2; N = 205) and obese (BMI ≥ 30 kg/m2; N = 69) groups. Their clinicopathological characteristics, outcomes, and survival rates were analyzed retrospectively.

Results

Obesity was associated with an increased rate of surgical complications such as wound infection (P < 0.001), fascial dehiscence (P = 0.023), and lymphoceles (P = 0.010). Furthermore, the hospital stay duration was significantly longer in the groups with obese patients compared to normal weight and overweight patients (normal weight: 22 days, overweight: 25 days, and obese: 33 days, respectively; P < 0.001). Multivariate analysis showed that recipient obesity (BMI ≥ 30) was an independent prognostic factor for delayed graft function (DGF) (OR 2.400; 95% CI, 1.365–4.219; P = 0.002) and postoperative surgical complications (OR 2.514; 95% CI, 1.230–5.136; P = 0.011). The mean death-censored graft survival was significantly lower in obese patients (normal weight: 16.3 ± 0.6 years, overweight: 16.3 ± 0.8 years, obese 10.8 ± 1.5 years, respectively; P = 0.001). However, when using the Cox proportional hazards model, the association between recipient obesity and death-censored renal graft failure disappeared, after adjustment for important covariates, whereas the principal independent predictors of graft loss were recipient diabetes mellitus and hypertension and kidneys from donors with expanded donor criteria.

Conclusion

In conclusion, obesity increases the risk of DGF and post-operative surgical complications after primary KT. Appropriate risk-adapted information concerning this must be provided to such patients before KT. Furthermore, obesity-typical concomitant diseases seem to negatively influence graft survival and need to be considered after the transplantation of obese patients.
Hinweise
Sebastian Rademacher and Robert Sucher  contributed equally to this work and share senior authorship.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
BMI
Body mass index
CIT
Cold ischemia time
CNI
Calcineurin inhibitor
CVA
Cerebrovascular accident
DD
Deceased donor
DGF
Delayed graft function
ECD
Expanded criteria donors
ESRD
End-stage renal disease
Ever
Everolimus
FFP
Fresh frozen plasma
GFR
Glomerular filtration rate
INF
Initial non-function
KT
Kidney transplantation
LD
Living donor
MMF
Mycophenolate mofetil
NODAT
New-onset diabetes after transplantation
POD
Post-operative day
POM
Post-operative month
PVD
Peripheral vascular disease
SD
Standard deviation
Siro
Sirolimus
WIT
Warm ischemia time

Introduction

Kidney transplantation (KT) is the treatment of choice in patients with end-stage renal disease (ESRD) and it improves both patient survival and recipients quality of life compared to chronic dialysis treatment [13].
Due to an aging society and changes in lifestyle – characterized by excessive calorie intake and a lack of physical activity – the percentage of overweight and obese patients has steadily increased in recent decades. According to the current Organisation for Economic Co-operation and Development (OECD) data, 60% of the population in Germany aged 15 years and older are overweight or obese [4]. Additionally, as obesity itself promotes ESRD, the proportion of obese renal transplant candidates is consequently increasing [59].
In general, surgery in obese patients is associated with a prolonged operative time and a higher risk for complications such as increased intraoperative blood loss and wound infections [1013]. And higher prevalence of comorbidities in obese patients—such as cardiovascular disease, diabetes mellitus and hypertension – could jeopardize the allograft. Therefore, transplantation in obese recipients is still discussed controversially.
Thus, the current study sought to analyze the short- and long-term outcomes of obese ESRD patients undergoing primary KT.

Patients and methods

Data collection and study population

Medical data from all adult patients (≥ 18 years of age) who underwent initial living or deceased donor kidney transplantation (KT) at the University Hospital of Leipzig between October 1993 and December 2017 were retrospectively analyzed. Our data source comprised a prospectively collected electronic database. Patients undergoing multi-organ (combined) transplants or re-transplants, underweight patients (BMI < 18.5 kg/m2), and patients with missing (incomplete) data were excluded from the study. Follow-up data were collected until March 2020.
The characteristics of the study population included donor and recipient age, gender, and body mass index (BMI, weight in kg/ height in m2), donor cause of death, duration of dialysis, time on the waiting list, and Criteria of Expanded Criteria Donors (ECD). Peri- and post-transplant data included information on the number of human leukocyte antigen (HLA) -A, B, and DR mismatches (0–6), last pretransplant panel reactive antibody (PRA) levels, cold (CIT) and warm ischemia time (WIT) of the grafts, duration of operation, as well as immunosuppressive therapy. CIT is defined as the time that the organ spent in cold preservation solution after removal from the donor, while WIT is the time from cross-clamping until cold perfusion, plus the time of implantation (organ out of ice until reperfusion).

Outcome measures

The outcome data included initial non-function (INF), biopsy-proven or clinically suspected episodes of acute rejection (in the first year after KT), delayed graft function (DGF), intra- and post-operative complications, date of graft failure, and patient death. INF was defined as dialysis dependence or creatinine clearance ≤ 20 mL/ min at three months post-transplant. Rejection episodes were histologically proved and DGF was defined as the need for dialysis in the first week following transplantation [14]. Post-operative complications occurring during the first three months after transplantation were analyzed. Complications included delayed wound healing, wound infection, urine leak, bleeding, development of hematoma, and lymphoceles. The Clavien-Dindo classification was used for morbidity assessment, and major morbidity was defined as being Clavien Dindo 3b or greater [15]. New-onset diabetes after transplantation (NODAT) was defined as the need for insulin or oral hypoglycemic drugs. Graft failure was defined as a return to dialysis or re-transplantation. Post-operative deaths included all deaths occurring within 30 days after surgery.

Body mass index

Recipient body mass index (BMI) was calculated based on the formula: weight (kg)/ [height (m)2], from height and weight recorded at the time of transplantation. Patients were classified as normal weight (BMI 18.5–24.9 kg/m2), overweight (BMI 25–29.9 kg/m2), or obese (BMI ≥ 30 kg/m2) according to guidelines of the World Health Organization (WHO) [16].

Glomerular filtration rate

Using serum creatinine levels, the estimated glomerular filtration rate (eGFR) was calculated using the Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation (mL/ min/ 1.73 m2 of standard body surface area (BSA)) [17]. To reduce errors induced by indexing the glomerular filtration rate for body surface area, the GFR was adjusted to the individual patient body surface area (eGFR x individual BSA [m2] / 1,73 m2 standard BSA = mL/ min) [18, 19].

Standard and expanded criteria donors

The standard criteria donor (SCD) was defined as a donor who is under 50 years of age and suffered brain death from any cause. Criteria of Expanded Criteria Donors (ECD) kidneys are sourced from donors over 60 years of age or donors between 50 and 59 years of age with at least two of the following three criteria: cerebrovascular death, arterial hypertension, and a donor serum creatinine level > 1.5 mg/dl [20].

Organ procurement and transplantation

The kidney grafts were procured according to the guidelines provided by Eurotransplant (ET) and transplanted into the iliacal fossa. Deceased donor kidneys were flushed in situ with cold HTK (histidine-tryptophan-ketoglutarate) solution and explanted. In living-related donation, kidneys were flushed with HTK after donor nephrectomy. For static cold storage, all grafts were immersed in HTK solution at 4 °C [21, 22]. The ureter was implanted into the bladder according to the Lich-Gregoir technique using a double J intra-ureteral splint [23, 24].

Immunosuppression

The initial immunosuppressive therapy comprised an induction therapy with the interleukin-2 receptor antagonists (daclizumab [withdrawn from the market in 2018] or basiliximab) or antithymocyte globulin, followed by triple maintenance immunosuppression comprising calcineurin inhibitors (tacrolimus or cyclosporine), and/or mTOR inhibitors (everolimus or sirolimus), antimetabolites (azathioprine or mycophenolate mofetil), and tapered steroids (prednisolone). A rapid steroid-tapering regimen was applied in all our patients, starting with 500 mg methylprednisolone intraoperatively to reach a dose of 25 mg prednisolone at the end of the first week after transplantation. Further reduction intended a daily maintenance dose of 5 mg. Whenever possible, steroids were rapidly withdrawn and discontinued at the end of the first year post-transplant.

Statistical analysis

For comparison between the groups, the appropriate statistical significance test, including Student’s t-test, the chi-squared test, analysis of variance (ANOVA), the Kruskal–Wallis test, and the Wilcoxon–Mann–Whitney test was used. Univariate and multivariate logistic regression analyses were used to evaluate the association between independent variables and binary outcomes of allograft function, and multivariate Cox proportional hazard analysis was applied to assess independent predictors of kidney graft failure. Prior to the regression analysis, post-operative complications were summarized into three groups: vascular (deep vein thrombosis, arterial or venous occlusion, secondary bleeding/hematoma), urological surgical (urine leakage), and surgical complications (wound infection, fascial dehiscence, lymphoceles). For the multivariate analyses, we used a forward stepwise regression model including only clinically relevant variables and those presenting P < 0.05 in univariate analysis. Survival rates were calculated using the Kaplan–Meier analysis, and the log-rank test was applied to test statistical significance. Graft survival was calculated as the time from initial transplant to graft failure (re-start of dialysis), uncensored for recipient death or censoring for death with a functioning graft. Patient survival was defined as the time from transplant to patient death, censoring for patients still alive at the time of analysis. If a recipient was alive or lost to follow-up at the time of the last contact, then survival time was censored at the time of the last contact. SPSS software, version 21.0 (SPSS Inc., Chicago, Illinois, USA) and Graphpad Prism software, version 9.2.0 (Graph-Pad Software Inc., La Jolla, CA) were used for statistical analysis and graphs. A P value < 0.05 was considered as statistically significant. Baseline data are presented as median values with the standard deviation (SD).

Results

Baseline characteristics

Between October 1993 and December 2017, 947 kidney transplants (KT) were performed in our department. A total of 578 patients could be included in the analysis, with 304 normal weight (52.6%), 205 overweight (35.5%), and 69 obese (11.9%) patients (Fig. 1).
The groups were similar in most of their baseline characteristics (Table 1). Median follow up was 5.8 ± 6.8 years. The proportion of obese patients significantly increased over the observation period (1993–2001: 7.0%, 2002–2009: 12.9%, and 2010–2017: 15.7%, respectively; P = 0.011). A higher BMI was associated with a significantly longer duration of surgery (P < 0.001); however, no significant differences between the groups were found for cold and warm ischemia time or intraoperative complications.
Table 1
Donor, recipient, and transplant characteristics
Variables
Body mass index
P-value
18.5–24.9 (N = 304)
25–29.9 (N = 205)
 ≥ 30 (N = 69)
Recipient
 Age, years
49.5 ± 14.2
55.9 ± 11.9
59.6 ± 12.2
 < 0.001
 Gender, male/ female (%)
182 (59.9)/ 122 (40.1)
135 (65.9)/ 70 (34.1)
46 (66.7)/ 23 (33.3)
0.304
 BMI, kg/m2
22.4 ± 1.6
27.1 ± 1.3
31.9 ± 2.4
 < 0.001
 Time on the waiting list, months
17.7 ± 28.6
18.4 ± 30.2
23.0 ± 30.3
0.574
 Dialysis duration, months
47.3 ± 35.5
46.4 ± 36.2
55.6 ± 33.6
0.763
Cause of ESRD (%)
 Glomerulonephritis
153 (50.0)
67 (32.7)
17 (24.6)
 < 0.001
 Non-glomerulonephritis, cystic kidney disease/ interstitial nephritis/ diabetes mellitus/ others/ unknown
47 (15.5)/ 28 (9.2)/ 3 (1.0)/ 50 (16.4)/ 24 (7.9)
39 (19.0)/ 26 (12.7)/ 16 (7.8)/ 42 (20.4)/ 15 (7.3)
15 (21.7)/ 7 (10.1)/ 10 (14.6) /16 (23.2)/ 4 (5.8)
 
Comorbidity (%)
 Diabetes mellitus
26 (8.6)
30 (14.6)
22 (31.9)
 < 0.001
 Hypertension
276 (90.8)
193 (94.1)
67 (97.1)
0.118
 Coronary disease
35 (11.5)
38 (18.5)
12 (17.4)
0.072
 PVD
11 (3.6)
16 (7.8)
6 (8.7)
0.071
Donor
 Age, years
51 ± 15.6
53 ± 17.3
56 ± 16.4
0.069
 Gender, male/ female (%)
165 (54.3)/ 139 (45.7)
114 (55.6)/ 91 (44.4)
41 (59.4)/ 28 (40.6)
0.737
 BMI, kg/m2
25.1 ± 4.3
25.0 ± 3.7
24.4 ± 3.0
0.200
 Organ quality, excellent/ good/ acceptable/ unknown (%)
14 (4.6)/ 216 (71.1)/ 10 (3.3)/ 64 (21.1)
17 (8.3)/ 136 (66.3)/ 11 (5.4)/ 41 (20.0)
5 (7.2)/ 50 (72.5)/ 4 (5.8)/ 10 (14.5)
0.329
Comorbidity (%)
 Diabetes mellitus
22 (7.2)
13 (6.3)
5 (7.2)
0.921
 Hypertension
83 (27.3)
71 (34.6)
25 (36.2)
0.129
 Donor type, LD/ SCD/ ECD (%)
62 (20.4)/ 146 (48.0)/ 96 (31.6)
26 (12.7)/ 100 (48.8)/ 79 (38.5)
5 (7.2)/ 32 (46.4)/ 32 (46.4)
0.014
Cause of death DD (%)
 CVA
125 (51.7)
92 (51.4)
35 (54.7)
0.571
 Non-CVA, anoxia/ ischemia/ polytrauma/ others
18 (7.4)/ 28 (11.6)/ 38 (15.7)/ 8 (3.3)
18 (10.1)/ 16 (8.9)/ 25 (14.0)/ 13 (7.3)
7 (10.9)/ 4 (6.3)/ 9 (14.1)/ 3 (4.7)
 
Transplant
 Transplant era (%)
   1993–2001/ 2002–2009/ 2010–2017
108 (35.5)/ 105 (34.5)/ 91 (29.9)
64 (31.2)/ 71 (34.6)/ 70 (34.1)
13 (18.8)/ 26 (37.7)/ 30 (43.5)
0.079
   HLA mismatches ≥ 3 (%)
143 (47.0)
108 (52.7)
40 (58.0)
0.271
   PRA (%)
50 (16.4)
29 (14.1)
10 (14.5)
0.753
   Gender mismatch
153 (50.3)
109 (53.29
31 (44.9)
0.462
   CIT, hours
11.5 ± 7.1
10.6 ± 6.6
11.4 ± 5.7
0.999
   WIT, minutes
40 ± 19.9
40 ± 15.8
45 ± 23.8
0.312
   Induction therapy, ATG/ ILR2-RA
15 (4.9)/ 101 (33.2)
7 (3.4)/ 68 (33.2)
2 (2.9)/ 26 (37.7)
0.582
 Intra-operative complications (%)
   Bleeding
11 (3.6)
10 (4.9)
3 (4.3)
0.780
   Thrombosis artery
8 (2.6)
6 (2.9)
3 (4.3)
0.748
   Thrombosis vein
4 (1.3)
2 (1.0)
1 (1.4)
0.925
   Hyperacute rejection
3 (1.0)
0
0
0.257
   Duration of surgery, minutes
172 ± 46.0
179 ± 53.2
194 ± 64.8
 < 0.001
 Immunosuppression (%)
   CNI, Tac/ CsA
170 (55.9)/ 128 (42.1)
122 (59.5)/ 75 (36.6)
47 (68.1)/ 22 (31.9)
0.400
   mTOR inhibitor, Ever/ Siro
1 (0.3)/ 8 (2.6)
0/ 4 (2.0)
1 (1.4)/ 1(1.4)
0.471
   CNI + mTOR inhibitor
9 (3.0)
4 (2.0)
2 (2.9)
0.788
   AM drug, AZA/ MMF
23 (7.6)/ 262 (86.2)
8 (3.9)/ 180 (87.8)
3 (4.3)/ 63 (91.3)
0.502
   Steroids, prednisolone
295 (96.7)
191 (93.2)
69 (100.0)
0.134
   Follow-up, years
7.6 ± 6.0
6.7 ± 5.9
5.5 ± 4.3
0.005
Data are shown as median ± SD
AM Antimetabolite, Aza Azathioprin, ATG Anti-thymocyte globulin, BMI Body mass index, CNI Calcineurin inhibitor, CsA Ciclosporin A, CVA Cerebrovascular accident, CIT Cold ischemia time, DD Deceased donor, ECD Expanded criteria donor, ESRD End-stage renal disease, Ever Everolimus, HLA Human leukocyte antigen-A, B, and DR, IL2-RA Interleukin-2 receptor antagonist, LD Living donor, MMF Mycofenolate mofetil, mTOR Mechanistic target of rapamycin, PVD Peripheral vascular disease, PRA Panel reactive antibody, SCD Standard criteria donor, Siro Sirolimus, WIT Warm ischemia time

Outcome

The analysis of post-operative outcome parameters is shown in Table 2. In the overall study population, 32 kidneys lost their function in the first three months (initial non-function, INF) (normal weight, over-weight, and obese KT recipients: 11, 16, and 5, respectively, P = 0.108), whereas permanent lack of graft function from the time of transplantation (primary non-function, PNF) was observed in five cases (normal weight, over-weight, and obese KT recipients: 2, 3, and 0, respectively, P = 0.447). No cases of PNF or INF were reported in kidney grafts after living donation.
Table 2
Post-operative outcome parameters and immunosuppression after deceased donor kidney transplantation
Variables
Body mass index
P-value
18.5–24.9 (N = 304)
25–29.9 (N = 205)
 ≥ 30 (N = 69)
Outcome Parameters
 Surgical
  RBC substitution (%)
83 (27.3)
55 (26.8)
17 (24.6)
0.903
  FFP substitution (%)
25 (8.2)
15 (7.3)
4 (5.8)
0.775
  Time on ICU, days
5 ± 5.0
5 ± 6.0
5 ± 14.6
0.079
   Post-operative complications (%)
   Deep vein thrombosis
3 (1.0)
4 (2.0)
2 (2.9)
0.435
   Occlusion or thrombosis
    Renal artery
5 (1.3)
4 (2.0)
2 (2.9)
0.631
    Renal vein
8 (2.6)
3 (1.5)
1 (1.4)
0.615
   Pulmonary embolism
2 (0.7)
1 (0.5)
0
0.788
   Secondary bleeding/ hematoma
62 (20.4)
39 (19.0)
16 (23.2)
0.754
   Wound infection
48 (15.8)
45 (22.0)
30 (43.5)
 < 0.001
   Fascial dehiscence
1 (0.3)
5 (2.4)
3 (4.3)
0.023
   Urine leakage
12 (4.0)
3 (1.5)
2 (2.9)
0.266
   Lymphocele
37 (12.2)
19 (9.3)
16 (23.2)
0.010
   Clavien-Dindo ≥ 3b (%)
67 (22.0)
51 (24.9)
26 (37.7)
0.025
 Renal
  INF (%)
11 (3.6)
16 (7.8)
5 (7.2)
0.108
  DGF (%)
72 (23.7)
47 (22.9)
32 (46.4)
 < 0.001
  Acute rejection, (%)
95 (31.3)
66 (32.2)
23 (33.3)
0.718
GFR (mL/ min)
   POD7
36.5 ± 42.5
45.5 ± 40.6
18.1 ± 28.3
0.002
   POD14
67.6 ± 46.8
62.0 ± 36.8
56.3 ± 35.6
0.041
   POM1
82.5 ± 36.5
76.7 ± 37.9
78.9 ± 35.9
0.160
   POM6
85.4 ± 32.3
80.9 ± 35.7
93.6 ± 33.9
0.704
 Metabolic
  NODAT (%)
15 (4.9)
12 (5.8)
8 (11.6)
0.110
 Hospitalisation, days
22 ± 16.1
25 ± 16.2
33 ± 21.5
 < 0.001
 Post-operative mortality 30 days (%)
3 (1.0)
3 (1.5)
0
0.579
 Post-operative mortality 90 days (%)
6 (2.0)
5 (2.4)
0
0.436
Data are shown as median ± SD
DGF delayed graft function, FFP fresh frozen plasma, GFR glomerular filtration rate, ICU intensive care unit, INF initial non-function, NODAT new-onset diabetes mellitus after transplantation, POD post-operative day, POM post-operative month, RBC red blood cells
Obese patients more frequently suffered from delayed graft function (DGF). These differences were most apparent in the subgroup of living donation (DGF normal weight, overweight, and obese, LD: 7 (11.3%), 2 (7.7%), and 3 (60.0%), respectively, P = 0.005; DD: 65 (26.9%), 45 (25.1%), and 29 (45.3%), respectively, P = 0.005). Fig. 2 shows glomerular filtration (GFR) rates among the groups within the first six months after KT.
The length of time spent in the intensive care unit after KT was comparable between the three groups (P = 0.079). However, the number of patients receiving ventilation post-transplant was significantly higher in the obese group (normal weight, over-weight, and obese KT recipients: 37 (12.2%), 36 (17.6%), and 16 (23.2%), respectively, P = 0.041), whereas the time of post-operative ventilation failed to show significance (normal weight, over-weight, and obese KT recipients: 2.0 ± 6.2 h, 2.0 ± 38.6 h, and 2.75 ± 33.1 h, respectively, P = 0.347).
Comparing Clavien-Dindo scores ≥ 3b, there was a significant difference in major complication rate between the groups (P = 0.025). Obesity was especially associated with an increased rate of surgical complications such as wound infection (P < 0.001), fascial dehiscence (P = 0.023), and lymphoceles (P = 0.010). Univariate analysis revealed that especially a recipient BMI ≥ 30 is significantly correlated with postoperative surgical complications and graft dysfunction (Table 3). This was underlined by a significantly longer hospital stay of obese patients compared to normal weight and overweight patients (normal weight, over-weight, and obese KT recipients: 22 days, 25 days, and 33 days, respectively; P < 0.001).
Table 3
Univariate analysis of recipient BMI and peri-operative kidney transplant outcome
Variables
Body mass index
18.5–24.9
25–29.9
 ≥ 30
Univariate analysis
Univariate analysis
OR
95% CI
P-value
OR
95% CI
P-value
Intraoperative complications
 Bleeding (1)
ref
1.366
0.569–3.278
0.485
1.211
0.329–4.461
0.774
 Thrombosis artery or vein (1)
ref
0.988
0.397–2.461
0.980
1.106
0.304–4.030
0.879
Duration of surgery (1)
ref
1.391
0.960–2.016
0.081
2.765
1.489–5.134
0.001
Ventilation time (1)
ref
1.524
0.871–2.665
0.140
2.510
1.236–5.097
0.011
Time on ICU (1)
ref
0.756
0.528–1.082
0.126
1.082
0.632–1.851
0.773
Postoperative complications
 Vascular (1)
ref
0.897
0.586–1.374
0.671
1.339
0.747–2.402
0.327
 Surgical (1)
ref
1.131
0.783–1.633
0.512
3.240
1.880–5.585
 < 0.001
 Urological surgical (1)
ref
0.361
0.101–1.297
0.118
0.726
0.159–3.322
0.680
 Clavien-Dindo ≥ 3 (%)
ref
1.171
0.772–1.777
0.457
2.148
1.225–3.734
0.008
 Hospitalisation (1)
ref
1.222
0.856–1.743
0.269
2.456
1.400–4.307
0.002
Renal outcome
 INF (1)
ref
2.232
1.014–4.914
0.046
2.092
0.702–6.234
0.185
 DGF (1)
ref
0.946
0.622–1.440
0.796
2.827
1.640–4.875
 < 0.001
 Acute rejection, all (2)
ref
1.121
0.861–1.458
0.397
1.494
0.861–2.171
0.035
 Graft failure (death-censored) (2)
ref
1.113
0.767–1.617
0.573
2.405
1.512–3.827
 < 0.001
NODAT (1)
ref
1.198
0.549–2.615
0.650
2.527
1.026–6.224
0.044
Prior to regression analysis, post-operative complications were summarized into three groups: vascular (deep vein thrombosis, arterial or venous occlusion, secondary bleeding/ hematoma), urological surgical (urine leakage), and surgical complications (wound infection, fascial dehiscence, lymphoceles).1, Univariate log regression; 2, univariate cox regression; 95% CI 95% confidence interval, DGF Delayed graft function, ICU Intensive care unit, INF Initial non-function, NODAT new-onset diabetes mellitus after transplantation, OR odds ratio, Ref Reference
In the group of normal-weight and overweight patients, there were five in-hospital deaths each, whereas no in-hospital deaths were reported in the obese group (P = 0.585). The causes of death included septic shock and multiple organ failure (N = 3), liver insufficiency (N = 1), and subdural hematoma (N = 1) in the normal weight group and septic shock (N = 1), endocarditis (N = 1), acute bleeding with cardiac arrest (N = 1) and acute heart failure (N = 2) in the overweight group, respectively.
In the overall study period, acute rejection was clinically suspected or histologically proven in 184 (31.8%) patients. In 149 cases, percutaneous kidney biopsies were performed and revealed acute rejection in 57 kidney allografts (38.3%), without showing any significant difference between the three groups (P = 0.918). However, an increase in the total number of treated rejection episodes per KT could be demonstrated among the groups, although these trends were also not statistically significant (normal weight: 8.1/ 100 graft years, overweight: 9.6/ 100 graft years, and obese: 17.7/ 100 graft years, respectively; P = 0.133) within the investigation period.

Graft and patient survival

Figure 3 shows Kaplan–Meier survival curves of grafts and patients after KT according to their BMI. The one-, three-, five- and ten-year cumulative death-censored graft survival rates were 92%, 88%, 83%, and 72%, respectively. The mean cumulative death-censored graft survival was 15.9 ± 0.5 years. Mean death-censored graft survival was significantly lower in obese patients (normal weight: 16.3 ± 0.6 years, overweight: 16.3 ± 0.8 years, obese 10.8 ± 1.5 years, respectively; P = 0.001). After ten years of follow-up, graft survival was 39% in the obese group compared with 78% in the normal weight and 73% in the overweight group.
The one-, three-, five- and ten-year cumulative patient survival rates were 96%, 92%, 89%, and 79%, respectively. The mean patient survival was 18.7 ± 0.5 years. The one-, three-, five- and ten-year survival rates, as well as the mean patient survival, were comparable between the groups (mean patient survival: normal weight: 19.2 ± 0.6 years, overweight: 17.6 ± 0.7 years, and obese: 15.8 ± 1.5 years, respectively; P = 0.186).
The causes of graft loss are summarized in Table 4. The main reasons for graft failure were acute or chronic rejection (33.1%) and chronic allograft nephropathy (22.3%). Among the three BMI groups, the percentage of graft losses due to infection was significantly enhanced in obese KT recipients (6 (8.1%), 5 (10.4%), 9 (31.0%); P = 0.006). No statistical difference could be observed between the three groups in regard to graft loss due to rejection (29 (39.2%), 13 (27.1%), 7 (24.1%); P = 0.193).
Table 4
Causes of renal graft loss according to recipient body mass index. Category numbers do not sum up due to overlap between causes of graft failure (normal weight: N = 2, obese: N = 1)
Variables
Body mass index
P-value
18.5–24.9 (N = 304)
25–29.9 (N = 205)
 ≥ 30 (N = 69)
 
Cause of graft loss (%)
 Death
23 (7.6)
13 (6.3)
4 (5.8)
0.125
 Graft failure
72 (23.7)
48 (23.4)
28 (40.6)
0.007
Cause of graft failure (%)
 Vascular
7 (9.5)
8 (16.7)
3 (10.3)
0.505
 Acute or chronic rejection
29 (39.2)
13 (27.1)
7 (24.1)
0.193
 Recurrence of renal disease
4 (5.4)
2 (4.2)
1 (3.4)
0.893
 Chronic allograft nephropathy
19 (25.7)
8 (16.7)
6 (20.7)
0.452
 Infection
6 (8.1)
5 (10.4)
9 (31.0)
0.006
 Other
9 (12.2)
12 (25.0)
3 (10.3)
0.130
In the multivariate regression analysis, obesity (BMI ≥ 30) remained an independent predictor of DGF (OR 2.400; 95% CI, 1.365–4.219; P = 0.002) and postoperative surgical complications (OR 2.514; 95% CI, 1.230–5.136; P = 0.011) (Table 5). However, after adjusting for important covariates, obesity failed to be an independent predictor of decreased graft survival or acute rejection. Independent predictors of graft loss were recipient diabetes mellitus and hypertension and kidneys from donors with expanded donor criteria. Expanded criteria donor was the only independent predictor of acute rejection (treated acute rejection (OR 1.448; 95% CI, 1.131–1.852; P = 0.003), biopsy-proven acute rejection (OR 1.919; 95% CI, 1.236–2.978; P = 0.004)).
Table 5
Multivariate regression analysis of predictors of delayed graft function, surgical post-operative complications, rejection, and kidney graft loss after kidney transplantation
A
Variables
Delayed graft function (1)
Surgical complications (1)
UVA
Multivariate analysis
UVA
Multivariate analysis
P-value
OR
95% CI
P-value
P-value
HR
95% CI
P-value
Recipient
 Age
0.001
1.535
1.003–2.350
0.048
0.040
NS
NS
NS
 BMI
 18.5–24.9 vs. 25–29.9
0.796
0.821
0.530–1.273
0.379
0.512
1.061
0.631–1.784
0.823
 18.5–24.9 vs. ≥ 30
 < 0.001
2.400
1.365–4.219
0.002
 < 0.001
2.441
1.182–5.038
0.016
 Diabetes mellitus
0.179
   
0.075
   
 Hypertension
0.512
   
0.388
   
 Coronary disease
0.043
NS
NS
NS
0.202
   
Donor
 Age
0.077
   
0.296
   
 Donor type, LD/ DD
0.002
2.118
1.091–4.112
0.027
0.071
   
 Cause of death, CVA/ non-CVA
0.960
   
0.457
   
 Diabetes mellitus
0.358
   
0.700
   
 Hypertension
0.013
NS
NS
NS
0.027
NS
NS
NS
 ECD
0.001
1.538
1.016–2.328
0.042
0.194
   
Transplant
 WIT
0.294
   
0.003
1.906
1.183–3.071
0.008
 Duration of surgery
0.017
NS
NS
NS
0.380
   
 Acute rejection
N/A
   
0.005
2.006
1.084–3.713
0.027
B
Variables
Rejection (treated) (2)
Graft failure (death-censored) (2)
UVA
Multivariate analysis
UVA
Multivariate analysis
P-value
HR
95% CI
P-value
P-value
HR
95% CI
P-value
Recipient
 Age
0.146
   
0.051
   
 BMI
  18.5–24.9 vs. 25–29.9
0.397
NS
NS
NS
0.573
NS
NS
NS
  18.5–24.9 vs. ≥ 30
0.035
NS
NS
NS
 < 0.001
NS
NS
NS
 Diabetes mellitus
0.034
NS
NS
NS
0.001
1.709
1.067–2.736
0.026
 Hypertension
0.053
   
 < 0.001
2.565
1.514–4.344
0.001
 Coronary disease
0.283
   
0.194
   
Donor
 Age
0.004
NS
NS
NS
 < 0.001
2.725
1.841–4.033
 < 0.001
 Donor type, LD/ DD
0.046
NS
NS
NS
0.004
NS
NS
NS
 Cause of death, CVA/ non-CVA
0.191
   
0.031
NS
NS
NS
 Diabetes mellitus
0.089
   
0.011
NS
NS
NS
 Hypertension
0.182
   
 < 0.001
NS
NS
NS
 ECD
0.004
1.448
1.131–1.852
0.003
 < 0.001
3.095
2.115–4.530
 < 0.001
Transplant
 WIT
0.333
   
0.497
   
 Duration of surgery
0.144
   
0.375
   
 Acute rejection
N/A
   
 < 0.001
2.709
1.830–4.011
 < 0.001
Following variables were tested in univariate analysis but failed to show significancy: cold ischemia time, delayed graft function; donor BMI, donor cause of death (CVA versus non-CVA), donor comorbidity diabetes mellitus, HLA, human leukocyte antigen-A, B, and DR (0–2 versus ≥ 3); initial immunosuppression (calcineurin inhibitor versus mTOR inihibitor), induction therapy, initial non-function organ quality (excellent–good versus acceptable–poor), panel reactive antibodies, peripheral vascular disease; transplant era. 1, Univariate log regression; 2, univariate cox regression; 95% CI 95% Confidence interval, BMI Body mass index, CVA Cerebrovascular accident, DD Deceased donor, ECD Expanded criteria donor, HR Hazard ratio, LD Living donor, NS Not significant, OR Odds ratio, UVA Univariate analysis, WIT Warm ischemia time

Discussion

The proportion of obese patients receiving kidney transplantation (KT) has been increasing over the last decades [79] and is subsequently becoming one of the leading challenges for transplant surgeons. In the following, we will address the effect of obesity on the short- and long-term outcomes after living and deceased donor KT.

Post-operative complications

In the current study, obese KT recipients were significantly more likely to experience surgical complications such as wound infections, fascial dehiscence, and lymphoceles, compared with normal weight and overweight KT recipients. In reviewing the literature, wound complications were among the most common post-operative complications in obese patients after KT [25], and in accordance with the present results, previous publications have demonstrated that lymphoceles occur more often in obese patients after KT [2628]. It is assumed that poor vascularization and the reduced immune response of fat tissue lead to poorer wound healing in this group of patients. Furthermore, a larger wound area and technical challenges due to more difficult exposure result in longer operation times and thus in higher probabilities of wound infection in obese patients [1013]. The length of hospital stays, which is a surrogate marker for post-operative complications, was also significantly prolonged in the group of obese patients. This is in line with the retrospective study from Gore et al. published in 2006 about 20,000 patients after living or deceased KT, which revealed an independent effect of recipient BMI on the length of hospital stay in the adjusted analysis [29].
Despite a higher rate of major postoperative complications, no post-operative deaths occurred in the group with obese kidney grafts recipients within the first three months after KT in our study cohort. Especially cardiovascular risk factors must be excluded prior to KT, as fatal cardiac events are a common cause of morbidity and death after KT in obese recipients [3033]. Therefore, in our department, all patients routinely receive a cardiac stress test and, if necessary, a cardiac catheterization before enlisting for KT.

Delayed graft function

Consistent with previous publications, we could identify recipient obesity as an independent risk factor of delayed graft function (DGF) in our cohort [25]. In a retrospective single-center study of 1,132 kidney transplant recipients, Weissenbacher et al. also showed that recipient BMI correlates with the incidence of DGF after deceased donor KT (recipient BMI: OR 1.087; 95% CI, 1.043–1.132; P < 0.0001) [34]. Additionally, a previously published retrospective multi-center study by Foucher et al. including 3,071 non-obese (BMI < 30) and 615 obese (BMI ≥ 30) recipients of kidney transplants confirmed this observation (recipient BMI: OR 1.89; 95% CI, 1.56–2.29; P < 0.0001) [33].
Although the pathogenesis of DGF has not yet been fully elucidated, it is thought to be a result of immunologically and ischemia-induced graft injury [14, 35]. In our study, neither the duration of surgery, nor the ischemia time of the graft reached statistical significance concerning DGF [29, 34, 36]. However, it should be noted that there is an additional risk of DGF in obese patients when receiving kidneys from cadaveric donation, which have an increased susceptibility to ischemic injury [29, 3537]. Therefore, the percentage of DGF after deceased donor KT is inherently higher compared to living donor KT, as the prevalence of DGF ranges from 4 to 10% in patients after living KT, and between 2 and 50% in kidneys from brain-dead donors [35, 38]. In our study, the rate of DGF was remarkably high in the subgroup of obese patients receiving kidneys after living donation (60%). However, due to the low number of patients in this group (N = 5), a conclusive statement is not possible and further studies with larger patient number are needed.
Furthermore, it should be noted that in obese KT recipients, renal function was only reduced in the early postoperative period after transplantation. Six months after KT, eGFR were comparable between the three study groups, indicating a good graft function even in KT recipients with a higher BMI.

Acute rejection episodes

A tendency towards a higher frequency of acute rejections per graft could be observed in the obese group of our patients. Meta-analysis showed that patients with a higher BMI have a significantly greater risk of acute rejection after KT [26]. Especially morbid obesity (BMI ≥ 35) demonstrated an association with acute rejection [29, 39]. A more difficult dosage adjustment, the maintenance of an appropriate level of immunosuppression, and a “state of chronic low-grade inflammation “ in obese patients could be reasons for the enhanced rate of acute rejection in this group [4042]. In our analysis, due to low patient numbers (BMI ≥ 35, N = 8), we could not evaluate the effects of an extreme BMI on outcomes after kidney transplantation. Further studies are necessary to analyze the serum levels of immunosuppressants in obese KT recipients and, in conjunction with those from acute graft rejection.

Graft survival

In our study, compared with normal weight or overweight KT recipients, graft survival in obese KT recipients was significantly reduced after five and ten years of observation. Several previous reports have shown that obesity itself confers a negative outcome on kidney graft survival [25, 31, 36, 39, 4345]. However, after adjusting for important covariates, higher BMI was not an independent predictor for decreased graft survival in our analysis. Hence, the reasons for the increased risk of graft failure in overweight transplant recipients are most probably multifactorial. No correlation between the transplant era and patient outcome could be found in our data set. A higher prevalence of comorbidities in obese patients – such as cardiovascular disease, diabetes mellitus, and hypertension – could jeopardize the allograft, and an imbalance between donor and recipient weight might lead to renal hyperfiltration and graft injury [43]. In our study, obese patients were significantly older compared with non-obese patients at the time of KT and showed higher frequencies of secondary diseases of which the prevalence of diabetes mellitus and hypertension had an independent influence on all-cause graft failure. Especially since a new-onset diabetes mellitus is more often seen in obese patients after KT [33, 46, 47], this underlines the importance of strict follow-up care with intensive control of diabetes and high blood pressure to prevent or delay kidney graft failure.

Limitations

There are some limiting factors in this study. First, the retrospective non-randomized design of the study and single-center effect should be mentioned. Second, the long investigation period restricted data evaluation, thereby making further controlled and prospective studies necessary. Third, we solely used BMI to define obesity in our study, which may not be an appropriate measure to characterize the nutritional status of a patient. Therefore, further studies, with a measurement of body fat distribution and muscle mass, and their association with the risk of morbidity and mortality in transplant recipients would be of interest. Fourth, the influence of weight loss or gain on the KT outcome and possibilities of bariatric surgery could be highlighted in future studies.

Conclusion

In conclusion, data from our center indicate that obese patients after KT seem to suffer from surgical complications and DGF more frequently than non-obese patients. Therefore, appropriate risk-adapted information must be provided to these patient groups before KT. Furthermore, although obesity itself may not directly impair the survival of the kidney graft, secondary diseases such as diabetes mellitus and hypertension must be taken into consideration in obese KT recipients as they seem to negatively affect long-term graft survival.

Acknowledgements

Not applicable

Declarations

The study was approved by the local ethical commission board from the University of Leipzig (AZ EK: 111–16-14032016). Written informed consent from any patient for data collection in a prospectively collected data base is available. However, written informed consent to the study was waived by the local Ethics Committee (Ethics Committee of the first affiliated University Hospital of Leipzig University) in view of the retrospective design of the study, accordingly the national and local guidelines such as the fact that all clinical/ laboratory measurements and procedures were part of the routine care.

Competing interests

The authors declare that they have no conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. 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 in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
2.
Zurück zum Zitat Sureshkumar KK, Patel BM, Markatos A, Nghiem DD, Marcus RJ. Quality of life after organ transplantation in type 1 diabetics with end-stage renal disease. Clin Transpl. 2006;20(1):19–25 PubMed PMID: 16556148.CrossRef Sureshkumar KK, Patel BM, Markatos A, Nghiem DD, Marcus RJ. Quality of life after organ transplantation in type 1 diabetics with end-stage renal disease. Clin Transpl. 2006;20(1):19–25 PubMed PMID: 16556148.CrossRef
5.
Zurück zum Zitat Tsujimoto T, Sairenchi T, Iso H, Irie F, Yamagishi K, Watanabe H, Tanaka K, Muto T, Ota H. The dose-response relationship between body mass index and the risk of incident stage ≥3 chronic kidney disease in a general japanese population: the Ibaraki prefectural health study (IPHS). J Epidemiol. 2014;24(6):444–51. https://doi.org/10.2188/jea.je20140028 Epub 2014 Jul 5. PMID: 24998954; PMCID: PMC4213218.CrossRefPubMed Tsujimoto T, Sairenchi T, Iso H, Irie F, Yamagishi K, Watanabe H, Tanaka K, Muto T, Ota H. The dose-response relationship between body mass index and the risk of incident stage ≥3 chronic kidney disease in a general japanese population: the Ibaraki prefectural health study (IPHS). J Epidemiol. 2014;24(6):444–51. https://​doi.​org/​10.​2188/​jea.​je20140028 Epub 2014 Jul 5. PMID: 24998954; PMCID: PMC4213218.CrossRefPubMed
11.
Zurück zum Zitat Tjeertes EK, Hoeks SE, Beks SB, Valentijn TM, Hoofwijk AG, Stolker RJ. Obesity–a risk factor for postoperative complications in general surgery? BMC Anesthesiol. 2015;15:155. https://doi.org/10.1186/s12871-015-0096-7 Tjeertes,ElkeEKM[correctedtoTjeertes,EKM];Hoeks,SanneSE[correctedtoHoeks,SE];Beks,SabineSBJC[correctedtoBeks,SBJ];Valentign,TabitaTM[correctedtoValentijn,TM];Hoofwijk,AntonAGM[correctedtoHoofwijk,AGM];Sto.PMID:26228844;PMCID:PMC4520073 .CrossRefPubMedPubMedCentral Tjeertes EK, Hoeks SE, Beks SB, Valentijn TM, Hoofwijk AG, Stolker RJ. Obesity–a risk factor for postoperative complications in general surgery? BMC Anesthesiol. 2015;15:155. https://​doi.​org/​10.​1186/​s12871-015-0096-7 Tjeertes,ElkeEKM[correctedtoTjeertes,EKM];Hoeks,SanneSE[correctedtoHoeks,SE];Beks,SabineSBJC[correctedtoBeks,SBJ];Valentign,TabitaTM[correctedtoValentijn,TM];Hoofwijk,AntonAGM[correctedtoHoofwijk,AGM];Sto.PMID:26228844;PMCID:PMC4520073 .CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Winfield RD, Reese S, Bochicchio K, Mazuski JE, Bochicchio GV. Obesity and the Risk for Surgical Site Infection in Abdominal Surgery. Am Surg. 2016;82(4):331–6 PMID: 27097626.CrossRef Winfield RD, Reese S, Bochicchio K, Mazuski JE, Bochicchio GV. Obesity and the Risk for Surgical Site Infection in Abdominal Surgery. Am Surg. 2016;82(4):331–6 PMID: 27097626.CrossRef
15.
Zurück zum Zitat Dindo D, Demartines N, Clavien PA. Classification of surgical complications: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg. 2004;240:205–13.CrossRef Dindo D, Demartines N, Clavien PA. Classification of surgical complications: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg. 2004;240:205–13.CrossRef
18.
Zurück zum Zitat Du Bois D, Du Bois EF. A formula to estimate the approximate surface area if height and weight be known. 1916. Nutrition. 1989;5(5):303–11 discussion 312–3. PMID: 2520314.PubMed Du Bois D, Du Bois EF. A formula to estimate the approximate surface area if height and weight be known. 1916. Nutrition. 1989;5(5):303–11 discussion 312–3. PMID: 2520314.PubMed
20.
Zurück zum Zitat Rao PS, Ojo A. The alphabet soup of kidney transplantation: SCD, DCD, ECD– fundamentals for the practicing nephrologist. Clin J Am Soc Nephrol. 2009;4(11):1827–31.CrossRef Rao PS, Ojo A. The alphabet soup of kidney transplantation: SCD, DCD, ECD– fundamentals for the practicing nephrologist. Clin J Am Soc Nephrol. 2009;4(11):1827–31.CrossRef
22.
Zurück zum Zitat Ladurner R, Steurer W. Technik der Multiorganentnahme. Viszeralchirurgie. 2004;39:439–42.CrossRef Ladurner R, Steurer W. Technik der Multiorganentnahme. Viszeralchirurgie. 2004;39:439–42.CrossRef
23.
Zurück zum Zitat Sollinger HW, Odorico JS, Becker YT, D’Alessandro AM, Pirsch JD. One thousand simultaneous pancreas-kidney transplants at a single center with 22-year follow-up. Ann Surg. 2009;250:618–30.CrossRef Sollinger HW, Odorico JS, Becker YT, D’Alessandro AM, Pirsch JD. One thousand simultaneous pancreas-kidney transplants at a single center with 22-year follow-up. Ann Surg. 2009;250:618–30.CrossRef
24.
Zurück zum Zitat Sollinger HW, Odorico JS, Knechtle SJ, D’Alessandro AM, Kalayoglu M, Pirsch JD. Experience with 500 simultaneous pancreas-kidney transplants. Ann Surg. 1998;228:284–96.CrossRef Sollinger HW, Odorico JS, Knechtle SJ, D’Alessandro AM, Kalayoglu M, Pirsch JD. Experience with 500 simultaneous pancreas-kidney transplants. Ann Surg. 1998;228:284–96.CrossRef
31.
Zurück zum Zitat Aziz F, Ramadorai A, Parajuli S, Garg N, Mohamed M, Mandelbrot DA, Foley DP, Garren M, Djamali A. Obesity: An Independent Predictor of Morbidity and Graft Loss after Kidney Transplantation. Am J Nephrol. 2020;51(8):615–23. https://doi.org/10.1159/000509105 Epub 2020 Jul 28 PMID: 32721967.CrossRefPubMed Aziz F, Ramadorai A, Parajuli S, Garg N, Mohamed M, Mandelbrot DA, Foley DP, Garren M, Djamali A. Obesity: An Independent Predictor of Morbidity and Graft Loss after Kidney Transplantation. Am J Nephrol. 2020;51(8):615–23. https://​doi.​org/​10.​1159/​000509105 Epub 2020 Jul 28 PMID: 32721967.CrossRefPubMed
33.
Zurück zum Zitat Foucher Y, Lorent M, Albano L, Roux S, Pernin V, Le Quintrec M, Legendre C, Buron F, Morelon E, Girerd S, Ladrière M, Glotz D, Lefaucher C, Kerleau C, Dantal J, Branchereau J, Giral M, DIVAT consortium. Renal transplantation outcomes in obese patients: a French cohort-based study. BMC Nephrol. 2021;22(1):79. https://doi.org/10.1186/s12882-021-02278-1 PMID: 33673808; PMCID: PMC7934368.CrossRefPubMedPubMedCentral Foucher Y, Lorent M, Albano L, Roux S, Pernin V, Le Quintrec M, Legendre C, Buron F, Morelon E, Girerd S, Ladrière M, Glotz D, Lefaucher C, Kerleau C, Dantal J, Branchereau J, Giral M, DIVAT consortium. Renal transplantation outcomes in obese patients: a French cohort-based study. BMC Nephrol. 2021;22(1):79. https://​doi.​org/​10.​1186/​s12882-021-02278-1 PMID: 33673808; PMCID: PMC7934368.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Yarlagadda SG, Coca SG, Formica RN Jr, Poggio ED, Parikh CR. Association between delayed graft function and allograft and patient survival: a systematic review and meta-analysis. Nephrol Dial Transplant. 2009;24(3):1039–47. https://doi.org/10.1093/ndt/gfn667 Epub 2008 Dec 22 PMID: 19103734.CrossRef Yarlagadda SG, Coca SG, Formica RN Jr, Poggio ED, Parikh CR. Association between delayed graft function and allograft and patient survival: a systematic review and meta-analysis. Nephrol Dial Transplant. 2009;24(3):1039–47. https://​doi.​org/​10.​1093/​ndt/​gfn667 Epub 2008 Dec 22 PMID: 19103734.CrossRef
40.
Zurück zum Zitat Shibata N, Hoshino N, Minouchi T, Yamaji A, Park K, Tomoyoshi T, Abe H, Kodama M. Relationship between area under the concentration versus time curve of cyclosporin A, creatinine clearance, hematocrit value, and other clinical factors in Japanese renal transplant patients. Int J Clin Pharmacol Ther. 1998;36(4):202–9 PMID: 9587046.PubMed Shibata N, Hoshino N, Minouchi T, Yamaji A, Park K, Tomoyoshi T, Abe H, Kodama M. Relationship between area under the concentration versus time curve of cyclosporin A, creatinine clearance, hematocrit value, and other clinical factors in Japanese renal transplant patients. Int J Clin Pharmacol Ther. 1998;36(4):202–9 PMID: 9587046.PubMed
Metadaten
Titel
Recipient obesity as a risk factor in kidney transplantation
verfasst von
Uwe Scheuermann
Jonas Babel
Uta-Carolin Pietsch
Antje Weimann
Orestis Lyros
Katrin Semmling
Hans-Michael Hau
Daniel Seehofer
Sebastian Rademacher
Robert Sucher
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
BMC Nephrology / Ausgabe 1/2022
Elektronische ISSN: 1471-2369
DOI
https://doi.org/10.1186/s12882-022-02668-z

Weitere Artikel der Ausgabe 1/2022

BMC Nephrology 1/2022 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.