Skip to main content
Erschienen in: BMC Infectious Diseases 1/2017

Open Access 01.12.2017 | Research article

Cytomegalovirus viraemia and mortality in renal transplant recipients in the era of antiviral prophylaxis. Lessons from the western Australian experience

verfasst von: Linda A. Selvey, Wai H. Lim, Peter Boan, Ramyasuda Swaminathan, Claudia Slimings, Amy E. Harrison, Aron Chakera

Erschienen in: BMC Infectious Diseases | Ausgabe 1/2017

Abstract

Background

Cytomegalovirus (CMV) establishes a lifelong infection that is efficiently controlled by the immune system; this infection can be reactivated in case of immunosuppression such as following solid organ transplantation. CMV viraemia has been associated with CMV disease, as well as increased mortality and allograft failure. Prophylactic antiviral medication is routinely given to renal transplant recipients, but reactivation during and following cessation of antiviral prophylaxis is known to occur. The aims of this study were to assess the incidence, timing and impact of CMV viraemia in renal transplant recipients and to determine the level of viraemia associated with adverse clinical outcomes.

Methods

Data from all adult (18 years and over) Western Australian renal transplant recipients transplanted between 1 January 2007 and 31 December 2012 were obtained from the Australia and New Zealand Dialysis and Transplant registry and were supplemented with data obtained from clinical records. Potential risk factors for detectable CMV viraemia (≥600 copies/ml) and all-cause mortality were assessed using univariable analysis and Cox Proportional Hazards Regression.

Results

There were 438 transplants performed on 435 recipients. The following factors increased the risk of CMV viraemia with viral loads ≥600 copies/ml: Donor positive/Recipient negative status; receiving a graft from a deceased donor; and receiving a graft from a donor aged 60 years and over. CMV viraemia with viral loads ≥656 copies/ml was a risk factor for death following renal transplantation, as was being aged 65 years and above at transplant, being Aboriginal and having vascular disease. Importantly 37% of the episodes of CMV viraemia with viral loads ≥656 copies/ml occurred while the patients were expected to be on CMV prophylaxis.

Conclusions

CMV viraemia (≥656 copies/ml) was associated with all-cause mortality in multivariable analysis, and CMV viraemia at ≥656 copies/ml commonly occurred during the period when renal transplant recipients were expected to be on antiviral prophylaxis. A greater vigilance in monitoring CMV levels if antiviral prophylaxis is stopped prematurely or poor patient compliance is suspected could protect some renal transplant recipients from adverse outcomes such as premature mortality.
Abkürzungen
aHR
adjusted hazard ratio
ANZDATA
Australia and New Zealand Dialysis and Transplant registry
CI
Confidence interval
CMV
Cytomegalovirus
D
Donor has negative CMV serology
D+
Donor has positive CMV serology
HLA
Human leukocyte antigen
HR
Hazard ratio
IQR
Interquartile range
NAT
Quantitative nucleic acid testing
PRA
Panel reactive antibody
R
Recipient has negative CMV serology prior to transplantation
R+
Recipient has positive CMV serology prior to transplantation
WA
Western Australia

Background

Cytomegalovirus (CMV) infection has long been recognised as a cause of morbidity and premature mortality amongst solid organ transplant recipients [1]. In Australia, the prevalence of CMV infection is estimated to be 57% among people up to 60 years of age [2]. The risks of CMV-related complications in kidney transplant recipients vary according to the serostatus of the donor (D) and recipient (R), with kidneys from CMV positive donors (D+) transplanted into CMV negative recipients (R) carrying the highest risk [1]. Reactivation of CMV in kidney transplant recipients may manifest across a clinical spectrum from asymptomatic viraemia to tissue-invasive disease [3]. Epidemiological studies have also linked CMV to chronic allograft injury, accelerated atherosclerosis and malignancy, potentially caused by direct viral cytopathic effects as well as immune modulation induced by the virus [46].
CMV prophylaxis with antivirals, such as ganciclovir or valganciclovir, has been in use for a number of years in Australia and elsewhere. The current Australian guidelines recommend 3 months of antiviral prophylaxis for D+/R+ and D/R+ recipients, 6 months for D+/R recipients, and none if both donor and recipient were negative for CMV [7]. CMV prophylaxis has been shown to reduce the risk of CMV disease and all-cause mortality in renal transplant recipients [8].
Despite the use of antiviral prophylaxis, CMV viraemia in transplant recipients remains a significant concern, with up to 35% of patients who were provided with prophylaxis becoming viraemic within 12 months of transplantation [1, 9]. In addition to the cost of prophylaxis and the risks of side-effects, in particular neutropenia, a concern is the development of late-onset disease (CMV viraemia occurring following cessation of prophylaxis), which is associated with allograft rejection [10] and increased mortality [11, 12].
Although a number of laboratory techniques exist for the detection of CMV viraemia, most centres across Australia are now performing quantitative nucleic acid testing (NAT). While very high levels of viraemia are typically associated with the presence of CMV syndrome or tissue-invasive disease, the clinical significance of lower-level viraemia remains unclear [13], and differences in NAT assays can lead to significant discrepancies in the quantification of viraemia between laboratories [14].
The aims of this study were to assess the incidence, timing and impact of CMV viraemia in renal transplant recipients and to determine the level of viraemia associated with adverse clinical outcomes.

Methods

Study population

All adult (age ≥ 18 years) patients who received a live or deceased donor kidney transplant in Perth, Western Australia (WA), between January 2007 and December 2012 were included in this study. Data were extracted from local health records and linked to data recorded in the Australia and New Zealand Dialysis and Transplant (ANZDATA) registry. If a patient received more than one kidney transplant during the study period, data regarding both grafts were extracted and both transplants were considered as separate grafts for analysis.
In WA, most patients received basiliximab induction and routine triple immunosuppression with a calcineurin inhibitor (CyA/tacrolimus), mycophenolate sodium or mycophenolate mofetil and oral prednisolone. Patients at lower risk of rejection did not receive either basiliximab or thymoglobulin induction and those at highest risk (around 10%) received thymoglobulin induction instead of basiliximab. Target tacrolimus trough levels are typically between 5 and 10 μg/L for the first 3 months, with the dose of prednisolone reduced to 5–10 mg daily by 3 months. Biopsy-proven T cell mediated rejection episodes were managed with intravenous methylprednisolone, with T cell depleting antibody being prescribed for steroid-resistant rejection episodes.
CMV prophylaxis with valganciclovir was administered for 100 days to R+ patients, 200 days to D+/R grafts, and no prophylaxis was given to D/R grafts. CMV testing at both centres during or after prophylaxis is usually performed on a monthly basis throughout the first year post-transplant. After that time, testing was done on an adhoc basis, usually only if there was a clinical indication. Valganciclovir at induction doses was routinely given following CMV reactivation at a viral load between 103 and 105 copies/mL. CMV treatment was routinely stopped after CMV was undetectable for two consecutive tests over at least a 1 month period.

Data collection

The following data were obtained from ANZDATA (to December 2013): age at transplantation; graft number; date of transplantation; date of birth; racial origin; weight and height at transplant; maximum panel reactive antibody (PRA) status; recipient smoking status; recipient CMV serology status; donor source; donor age; human leukocyte antigen (HLA) mismatches; occurrence of acute rejection (including rejection type, severity and treatment); induction therapy (interleukin-2 receptor antibody or T cell depleting antibody); immunosuppression prescribed at 0, 3, 6 months and at each year post-transplant; comorbidities of cardiovascular disease, chronic lung disease, diabetes or cancer (post-transplant or recurrent); whether or not graft failure occurred and the date of failure; and whether or not the patient died (to December 2014) and the date of death. Information on treatment for CMV viraemia was not available.
Donor CMV serology status was obtained from individual patient charts at the two transplant hospitals. Recipient CMV serology status was confirmed from individual patient records and where differences between patient records and ANZDATA were identified, the patient record information was used. Any data missing from the ANZDATA dataset were also obtained from patient records where possible.
CMV DNA plasma viral load testing was performed at PathWest Laboratory Medicine WA using the COBAS Amplicor CMV MONITOR assay (Roche Diagnostics, Branchburg, N.J., USA) from January 2007 to 12 May 2011, which has a linear range of 6 × 102 to 1 × 105 copies/mL (not referenced to the international CMV quantitative standard, so not reportable in IU/mL). From 13 May 2011 the Abbott RealTime CMV assay (Abbott Molecular Inc., Des Plaines, IL, USA) on the m2000 RealTime platform was introduced, which has a linear range of 2 × 101 to 1 × 108 copies/mL (multiplication of copies/mL by 1.56 converts the result to IU/mL according to the WHO international CMV quantitative standard). We obtained testing data (date of test, test result) up to December 2014 inclusive and CMV results were reported in copies/ml.

Data analysis

Incidence rates and 95% confidence intervals were calculated for all variables of interest for the outcomes: CMV viraemia with greater than or equal to 600 copies/ml; and patient death prior to 31 December 2014. The start time was the date of the first recorded transplant for calculating the incidence of CMV viraemia; and the date of the most recent recorded transplant for calculating the incidence of patient death prior to 2014. When calculating incidence for time-dependent variables, the start time was the time of transplant for the period when the variable equalled zero. If and when the categorical variables became greater than zero, this was the start time for incidence. Patient days prior to that time were counted as patient days for that variable being zero. The Exact method was used to estimate p-values for the significance of effect for binomial variables and univariable Cox Proportional Hazards analysis was used to estimate p-values for categorical variables with more than two possible values. Time-dependent variables, such as CMV, were fitted as time-dependent variables in the univariable Cox Proportional Hazards models. Age at transplantation was categorised to <65 and ≥65 on the basis of 65 being an age cut-off for increased risk of death and adverse outcomes in other studies [1517]. The cut-off for categorisation of CMV levels was set at 600 copies/ml on the basis that 600 copies/ml was the limit of detection for the less sensitive Roche COBAS Amplicor assay. The cut-off for categorisation of donor age was set at 60 years of age, and for categorisation of the total number of HLA mismatches the cut-off was set at two. Patient death outcome data were censored as of December 2014 and CMV viraemia outcome data were censored as of December 2013. The earlier censoring of CMV viraemia outcome data was because information about rejection and immunosuppression was only available up to December 2013.
Multivariable survival analysis was performed using Cox Proportional Hazards regression. Tied failures were calculated using the Breslow method for ties. Robust standard errors were used to estimate p values and 95% confidence intervals. Covariates with a p-value of 0.100 or less in the univariable models or covariates previously reported to correlate with study outcomes were included in the initial models. Variables were excluded from the model if their p-values for their adjusted hazard ratio was >0.05. For the model analysing CMV viraemia as the outcome, immunosuppression was categorised into three groups: calcineurin inhibitors (cyclosporin A or tacrolimus); antimetabolites (azathioprine, mycophenolate mofetil, or mycophenolate sodium); and mammalian target of rapamycin (mTOR) inhibitors (everolimus or sirolimus), and fitted as time-dependent covariates for each time period for which immunosuppression data were recorded in ANZDATA (3 months, 6 months, 1, 2, 3, 4, and 5 years). Anti-T lymphocyte antibody treatment after transplant was fitted as a time-dependant variable and was compared to having no treatment or treatment at or before transplantation. All rejection episodes were biopsy-proven with severe rejection defined as any vascular rejection or any severe humoral, glomerular or cellular rejection. Rejection variables were also fitted as time-dependent covariates. The following variables were included in the initial model for CMV viraemia: recipient age (categorised at 65 years), sex, donor age (categorised at 60 years), donor/recipient CMV status, immunosuppression, rejection, severe rejection, numbers of HLA mismatches (categorised as two or less or greater than two), and donor status (deceased or live). For the model examining death as the outcome, CMV status was fitted as a time-dependent covariate as one or more episodes (combined). The variables included in the initial model for death as an outcome were age (categorised at 65 years), sex, donor age (categorised at 60 years), donor status (deceased or live), rejection, severe rejection, CMV status, Aboriginality, smoking status, graft failure and the following comorbidities: diabetes; cerebrovascular disease, any cardiovascular disease other than cerebrovascular disease in patients with no cerebrovascular disease; chronic lung disease and cancer. The proportional hazards assumption was tested for all variables in every final model based on the analysis of Schoenfeld residuals.
A Kaplan-Meier plot showing death by CMV viral load was generated with CMV viral load being fitted as a time-dependent variable.
Data analysis was performed using STATA v14 (StataCorp LP, College Station, Texas, USA).

Results

There were 438 renal transplants performed in 435 recipients (with three patients receiving a second transplant in the study period) in WA from January 2007 to December 2012 and all cases were included in the analysis. The median age at transplant was 51 years (range 18–80).

Risk factors associated with CMV viraemia

One hundred and thirty four (30.6%) patients had at least one episode of detectable viraemia during the study period. Of these, 74 (55.2%) patients had at least one episode of viraemia at 600 copies/ml or more. Eleven (14.9%) of the 74 patients had two episodes of CMV viraemia at 600 copies/ml or more with intervening periods of no CMV DNA being detected. One patient had a single CMV viraemia episode of 600 copies/ml or more before transplantation. This viraemic episode was excluded, leaving in the analysis 84 CMV viraemia episodes of 600 copies/ml or more from 73 patients over 461,961 patient days. The incidence of CMV viraemia of 600 copies/ml or more was 18.2 per 100,000 person days (95%CI 14.7–22.5). Forty-six patients died before 31 December 2013 (Fig. 1). There were three patients who developed CMV viraemia after graft failure and had no record of a subsequent transplant. These patients were excluded from the univariable analysis, and the CMV episodes were excluded from the Cox Proportional Hazards analysis as they were censored at graft failure.
Table 1 presents the numbers and rates of CMV viraemia at 600 copies/ml or more stratified by covariate categories. Being 65 years or above at the time of transplant, having D+/R status, having at least one episode of severe rejection, and having a transplant from a deceased donor or a donor aged 60 years and over were all significantly associated with having CMV viraemia in univariable analysis (Table 1).
Table 1
CMV proportions and rates by covariates
 
Total n
Number with CMV (%)
CMV rate per 100,000 patient days (95% CI)
p-value
Total
435
70 (16.1)
18.2 (14.7, 22.5)
 
Sex
 Female
164
27 (16.5)
19.1 (13.6, 26.7)
0.893
 Male
272
43 (15.9)
17.6 (13.4, 23.3)
 
Age 65 and over
  < 65
396
58 (14.7)
16.4 (13.0, 20.7)
0.019
  ≥ 65
39
12 (30.8)
46.1 (26.8, 79.4)
 
Transplant Hospital
 SCGH
232
33 (14.2)
14.60 (10.58, 20.16)
0.296
 RPH
206
40 (19.4)
22.53 (16.93, 29.99)
 
Index graft history
 First graft
384
64 (16.7)
18.8 (15.0, 23.5)
0.425
 Had previous graft(s)
51
6 (11.8)
14.1 (7.0, 28.2)
 
Number of grafts during follow up
 Single graft
432
69 (16.0)
18.0 (14.5, 22.3)
0.410
 Two grafts
3
1 (33.3)
191.2 (26.9, 1357.4)
 
Donor & recipient CMV status (n = 430)
 DR
49
0 (0)
0
<0.001
 DR+
115
13 (11.3)
11.0 (6.5, 18.6)
 
 D+R
68
25 (36.8)
56.8 (40.0, 80.8)
 
 D+R+
195
32 (16.4)
18.9 (13.8, 25.9)
 
HLA mismatch (index graft) (n = 432)
  < 2 mismatches
58
5 (8.6)
8.3 (3.7, 18.5)
0.153
  ≥ 2 mismatches
371
65 (17.5)
20.2 (16.2, 25.2)
 
Antibody treatment
 None or before or at transplant only
394
59 (30.3)
15.0 (8.5, 26.5)
 
 Post-transplant
33
11 (33.3)
18.8 (15.0, 23.7)
0.687
Rejection (any)
 No
342
51 (14.9)
16.3 (12.6, 21.1)
0.205
 Yes
93
19 (20.4)
35.3 (16.6, 35.3)
 
Rejection (severe)
 No
407
63 (15.5)
16.7 (13.3, 21.1)
0.002
 Yes
28
7 (25.0)
38.3 (21.7, 67.4)
 
Aboriginal
 No
386
63 (16.3)
18.3 (14.6, 22.9)
0.838
 Yes
49
7 (14.3)
17.6 (9.2, 33.8)
 
Donor aged >60
 No
317
40 (12.6)
12.9 (9.6, 17.2)
0.002
 Yes
118
30 (25.2)
36.4 (26.5, 50.0)
 
Deceased donor
 No
192
25 (13.0)
11.4 (7.8, 16.8)
0.017
 Yes
244
45 (18.5)
24.8 (19.2, 32.1)
 

Multivariable analysis

For the Cox Proportional Hazards regression analysis, the three CMV episodes after transplant failure were excluded as the data were censored at transplant failure. CMV-negative recipients receiving grafts from CMV-negative donors were also excluded from the analysis, as none of these participants developed CMV viraemia. All three patients with a second graft during the follow up period exited the survival model at the failure date of the first graft. Having D+/R CMV status was associated with a five-fold increased risk of CMV viraemia of 600 copies/ml or more. Receiving a graft from a donor aged 60 or more, and having a graft from a deceased donor, were also significantly associated with CMV viraemia of 600 copies/ml or more. Immunosuppressive regimens that included an mTOR inhibitor (either everolimus or sirolimus) resulted in a 70% decreased risk of a CMV viraemic episode (as a time-dependent covariate), although this was not statistically significant (p = 0.052) (Table 2).
Table 2
Final Cox Proportional Hazards regression model for CMV incidence (with mTOR inhibitor fitted as a time dependent variable)
Variable
Adjusted Hazard Ratio (95% CI)
p-value
Recipient aged ≥65 at transplant
1.85 (0.96, 3.58)
0.067
Donor aged ≥60
2.24 (1.36, 3.67)
0.001
Deceased donor
2.03 (1.18, 3.50)
0.011
Donor/Recipient CMV status
 D/R+ (reference)
 D+/R
5.44 (2.49, 11.89)
<0.001
 D+/R+
1.73 (0.84, 3.56)
0.138
mTOR inhibitor
0.31 (0.09, 1.02)
0.054

Association between CMV viraemia and all-cause mortality

Of the 438 transplant recipients, 55 (12.6%) died before 31 December 2014. Having at least one episode of CMV detectable at 600 copies/ml or greater, being aged 65 or over at the time of transplant, receiving a graft from a deceased donor, lung disease, cerebrovascular disease, and Aboriginal race were all predictors of death in univariable analysis (Table 3).
Table 3
Univariable analysis of potential predictors of death amongst transplant recipients (n = 438)
Variable
Number
Number of deaths (%)
Death rate per 100,000 patient days (95% CI)
p-value
All cases
438
55 (12.6)
7.9 (6.0, 10.2)
 
Transplant Hospital
 SCGH
232
29 (12.5)
7.9 (5.5, 11.3)
1.0
 RPH
206
26 (12.6)
7.8 (5.3, 11.5)
 
CMV viral load of ≥600 copies/ml
 No
364
35 (9.6)
5.8 (4.2, 8.0)
<0.001
 Yes
74
20 (27.0)
21.0 (13.6, 32.6)
 
CMV viral load (copies/ml)
  < 600
364
35 (9.6)
5.8 (4.2, 8.0)
 
 600–4999
42
10 (23.8)
18.7 (9.7, 35.9)
<0.001
  ≥ 5000
32
10 (31.3)
23.5 (13.0, 42.4)
0.001
Sex
 Female
166
25 (15.1)
9.2 (6.2, 13.7)
0.30
 Male
272
30 (11.0)
7.0 (4.9, 10.0)
 
Deceased donor
 No
192
17 (8.9)
5.2 (3.2, 8.3)
0.02
 Yes
246
38 (15.5)
10.2 (7.4, 14.0)
 
Donor aged ≥60
 No
317
35 (11.0)
6.8 (4.9, 9.5)
0.10
 Yes
121
20 (16.5)
10.8 (7.0, 16.8)
 
Graft failure
 No
390
47 (12.1)
7.6 (5.7, 10.1)
0.51
 Yes
48
8 (16.7)
9.7 (4.9, 19.4)
 
Age ≥ 65 at transplant
 No
399
42 (10.5)
6.5 (4.8, 8.8)
<0.001
 Yes
39
13 (33.3)
24.2 (14.1, 41.7)
 
Lung Disease
 No
378
41 (10.9)
6.8 (5.0, 9.2)
0.02
 Yes
60
14 (23.3)
14.6 (8.6, 24.6)
 
Cerebrovascular disease
 No
407
41 (10.1)
6.2 (4.6, 8.4)
<0.001
 Yes
31
14 (45.1)
34.5 (20.4, 58.3)
<0.001
Vascular disease other than cerebrovascular
 No
104
36 (10.8)
6.7 (4.8, 9.3)
0.05
 Yes
334
19 (18.3)
11.7 (7.5, 18.4)
 
Diabetes
 No
293
35 (12.0)
7.5 (5.4, 10.4)
0.61
 Yes
145
20 (13.8)
8.6 (5.6, 13.3)
 
HLA mismatch ≥2
 No
373
6 (10.2)
5.9 (2.6, 13.1)
0.45
 Yes
59
49 (13.1)
8.3 (6.2, 10.9)
 
Maximum Panel Reactive Antibody level ≥ 50%
 No
366
46 (12.6)
7.8 (5.8, 10.4)
0.62
 Yes
61
9 (14.8)
9.2 (4.8, 17.7)
 
Any cancer diagnosis
 No
352
42 (11.9)
7.5 (5.5, 10.2)
0.51
 Yes
86
13 (15.1)
9.2 (5.3, 15.8)
 
Aboriginal
 No
387
40 (10.3)
6.4 (4.7, 8.8)
0.001
 Yes
51
15 (29.4)
19.3 (11.6, 32.0)
 
Any rejection
 No
302
37 (12.3)
7.9 (5.7, 10.9)
0.99
 Yes
136
18 (13.2)
7.8 (4.9, 12.4)
 
Severe rejection
 No
404
49 (12.1)
7.7 (5.8, 10.1)
0.54
 Yes
34
6 (17.7)
9.9 (4.4, 21.9)
 
Healthy weight (BMI 19–25) at transplant
 No
272
35 (12.9)
8.3 (5.9, 11.5)
0.78
 Yes
159
20 (12.6)
7.6 (4.9, 11.8)
 
Current smoker
 No
398
48 (12.1)
7.6 (5.7, 10.1)
0.45
 Yes
40
7 (17.5)
10.2 (4.9, 21.5)
 

Sensitivity analysis for CMV viraemia and mortality

We next sought to examine the relationship between a number of thresholds for CMV viral load, the duration of viraemia and all-cause mortality. There were 134 (30.6%) patients who had at least one episode of detectable CMV viraemia. The median CMV viral load among these cases was 656 copies/ml (IQR 68, 4590), and levels ranged from 2.6 to 5,710,000 copies/ml. Using the quartile values as cut-offs, the CMV viral load was categorised as <68, 68–655, 656–4589 and 4590+ copies/ml. A Cox Regression model showed a significant HR for death only for those cases with a CMV viral load of ≥656 copies/ml (Table 4). Kaplan Meier survival plots for death by CMV viral load are shown in Fig. 2.
Table 4
Cox Proportional Hazards Regression analysis for death by CMV level
Variable
Adjusted Hazard Ratio (95% CI)
p-value
CMV viral load <68 copies/ml (reference)
-
-
CMV viral load 68–655 copies/ml
0.63 (0.15, 2.62)
0.52
CMV viral load 656–4589 copies/ml
2.35 (1.12, 4.91)
<0.001
CMV viral load ≥4590 copies/ml
3.19 (1.61, 6.31)
0.001

Multivariable analysis for factors associated with all cause mortality

Using Cox Proportional Hazards regression analysis, having at least one episode of CMV viraemia with viral loads of 656 copies/ml or more, Aboriginal race, aged 65 or more at transplant, having cerebrovascular disease, and having any other vascular disease were all associated with death (Table 5). An interaction term of CMV with age was not significant and were therefore not included in the final model.
Table 5
Final Cox Proportional Hazards regression model for death
Variable
Adjusted Hazard Ratio (95% CI)
p-value
CMV viral load
 Less than 656 copies/ml (reference)
-
 
 656–4590 copies/ml
3.25 (1.42, 7.44)
0.007
  ≥ 4590 copies/ml
3.94 (1.75, 8.89)
0.012
Aboriginal
3.39 (1.74, 6.63)
<0.001
Age ≥ 65 at transplant
2.39 (1.10, 5.18)
0.027
Cerebrovascular disease
4.21 (1.81, 9.78)
0.001
Other vascular disease
2.21 (1.19, 4.09)
0.012

Multivariable analysis for factors associated with death with a functioning graft

Of the 55 patients who died, 47 died with a functioning graft. A new Cox Proportional Hazards regression model was generated censoring at graft failure the eight cases who died after graft failure. In this model, having at least one episode of CMV viraemia with levels of 656–4589 copies/ml (aHR 3.36, 95% CI 1.49, 7.55, p = 0.003), having at least one episode of CMV viraemia with levels of 4590 copies/ml or more (aHR 3.96, 95% CI 1.85, 8.49, p < 0.001), Aboriginal race (aHR 3.33, 95% CI 1.74, 6.36, p < 0.001), aged ≥65 at transplant (aHR 2.38, 95% CI 1.11, 5.13, p = 0.026), cerebrovascular disease (aHR 4.24, 95% CI 1.87, 9.66, p = 0.001), and other vascular disease (aHR 2.23, 95% CI 1.22, 4.08, p = 0.009) were associated with death with a functioning graft.

CMV duration

The duration of CMV viraemia was not associated with death. The incidence of death from most recent transplant was 5.8 (95% CI 4.2, 8.1) for patients with no episode of CMV viraemia of 656 copies/ml or more; 20.9 (95% CI 11.2, 38.8) for patients with CMV viraemia of 656 copies/ml or more for a duration of up to 7 days; and 21.0 (95% CI 11.3, 39.1) for patients with CMV viraemia of 656 copies/ml or more for a duration of at least 7 days. Further analysis comparing death with duration of CMV viraemia with a threshold of 14 days and with 28 days also did not show an association between duration of CMV viraemia and death (data not shown).

Onset of CMV viraemia

There were 73 patients who had at least one episode of CMV viraemia of 656 copies/ml or greater post-transplant. Eleven of these patients had two episodes of CMV viraemia of 656 copies/ml or greater (a total of 84 episodes in 73 patients). Of those viraemic episodes, 31 (37%) occurred during the period when the patient was expected to be on CMV prophylaxis.

Discussion

In this contemporary group of kidney transplant recipients, CMV viraemia with a threshold of at least 656 copies/ml was associated with over a three-fold greater risk of all-cause mortality and death with functioning graft. An important observation is that 37% of CMV viraemia of at least 656 copies/ml occurred during the period of presumed antiviral prophylaxis suggesting that close monitoring of CMV viraemia is warranted should patients cease antiviral prophylaxis prematurely or if non-compliance is suspected. Our finding that CMV viraemia of relatively low levels (≥656 copies/ml) was also associated with death also suggests vigilance with CMV treatment when CMV viraemia reaches these levels. Although we had expected that increasing duration of CMV viraemia would be associated with mortality, this was not apparent in this study. However, the inconsistent timing of CMV testing may have modified the association between duration of CMV viraemia and outcome. Our study builds on the work of previous investigators [11, 12, 1822], by being conducted at a time when all eligible renal transplant recipients were prescribed CMV prophylaxis, and by considering risk factors and consequences of CMV viraemia, rather than CMV disease.
Given that having CMV activation at viral loads of 656 copies/ml or greater is associated with death, our finding that 37% of the episodes of CMV activation at this level occurred while the patient was expected to be on CMV prophylaxis is of great concern. We were unable to determine whether this was due to a lack of compliance with CMV prophylaxis, or whether prophylaxis was withdrawn due to intolerable side effects. Our findings do highlight the critical importance of monitoring CMV viral loads during periods when antiviral prophylaxis has been prematurely ceased, and the provision of fairly aggressive CMV treatment should levels rise over 600 copies/ml. They also stress the importance of patient compliance with CMV prophylaxis and of ensuring dose adjustment of prophylaxis according to the patient’s glomerular filtration rate.
While the latest CMV assays are extremely sensitive and are able to detect low-level viraemia, it is unknown whether low-level viraemia below the threshold of earlier assays is clinically significant. Only levels of at least 656 copies/ml were associated with mortality in our analysis. While other studies have shown an association between acute rejection and risk of CMV viraemia, it was unclear whether the association between rejection and CMV viraemia was related to the increased dose of immunosuppression or a direct effect of the rejection process [22].
The association between Aboriginal status and death following transplantation has been previously described. Due to a range of factors, Aboriginal people are more likely to develop chronic renal failure and they have poorer outcomes than non-Aboriginal Australians post-transplant [23].
The association between the use of mTOR inhibitors and reduced rates of viral infections has been shown in several studies, including a recent meta-analysis in kidney transplant recipients [24, 25]. Although the use of mTOR inhibitors was associated with a reduced risk of CMV viraemia in the unadjusted model, this association was no longer present in the adjusted model, which may suggest a type II error or that reduced intensity of immunosuppression (as a result of eliminating calcineurin inhibitors) may be a more important factor in reducing the risk of CMV viraemia.
Receiving a graft from a donor aged 60 years and older has previously been found to be associated with late-onset CMV disease in D+/R patients [18]. To our knowledge, there are no other studies finding an association between receiving a graft from a deceased donor and CMV viraemia/disease. An interaction term between the variables, donor aged ≥60 years and deceased donor, was not significant in the model. While we did not find that the number of HLA mismatches between donor and recipient was associated with CMV viraemia, our finding may also be related to a reduced level of immunological matching between recipients and deceased donors compared to between recipients and live donors, therefore increasing the level of immunosuppression required to prevent rejection.
It is known that a high and rapid increase of plasma CMV viral load is associated with a greater likelihood of invasive CMV disease [13], however, CMV disease can occur even with only low levels of viral DNA detectable in plasma, as described with gastrointestinal and retinal disease [26]. There is no recognised cut-off for the commencement of antiviral therapy; many earlier studies were hampered by use of assays with no reference to the WHO international quantitative standard [13]. One recent study of CMV seropositive kidney, heart and liver transplant recipients found a high predictive value for CMV disease in those with CMV plasma viral load of 3983 IU/mL or more compared with lower levels [27]. Lower CMV viral loads not associated with a risk of invasive CMV disease may still be associated with the indirect effects of CMV. However, with a poor understanding of how antiviral therapy alters the indirect effects of CMV and the association of CMV viral loads with indirect CMV effects, there are no definite recommendations regarding antiviral therapy for this indication [28, 29].
There are several strengths and limitations to our study. This is one of the largest studies to evaluate the association between CMV viraemia and clinical outcomes in the era where antiviral prophylaxis is standard practice. Even though this is not a small study, our study may have had insufficient power to examine the association between CMV viraemia and several outcomes including death due to different causes. ANZDATA and patient records do not document the duration and dose of CMV prophylaxis and there is limited recording of patient compliance. Another limitation of our study is that the results in copies/mL for different CMV viral load assays used in this study are not directly comparable without being able to report IU/mL for both assays [13]. For the COBAS Amplicor assay the only available data relating copies/mL to IU/mL by the CMV international quantitative standard is provided by indirect comparison to the COBAS Ampliprep assay [30].

Conclusions

We found that CMV viraemia (at 656 copies/ml or greater) was associated with over a three-fold increased risk of all-cause mortality amongst renal transplant recipients. An association between CMV disease and all-cause mortality has been previously shown, but less frequently for CMV viraemia alone [1, 11, 18, 20, 22, 31, 32], although these findings are not consistent across all studies [12]. Despite the routine use of antiviral prophylaxis after kidney transplantation in R+ and D+/R- recipients, 37% of recipients experienced CMV viraemia during the recommended period for antiviral prophylaxis. Clinicians should be cognisant of the need to monitor and identify CMV viraemia when prophylaxis is prematurely ceased or non-compliance is suspected. Further examination of the association between duration of CMV viraemia and patient outcomes in larger cohorts is required. While some cases of CMV disease were recorded in the ANZDATA, there is not a specific data field where this can be documented and it is likely that this was significantly under-reported. Therefore, we have not assessed the relationship between CMV disease and transplant outcomes or factors associated with CMV disease. We were unable to determine whether or not our findings reflect a true impact of CMV viraemia on mortality, or whether CMV viraemia is a confounder for other, unmeasured factors associated with death [32].

Acknowledgements

The authors would like to gratefully acknowledge the substantial contributions of the entire Australian and New Zealand nephrology community in Western Australia (physicians, surgeons, database managers, nurses, renal operators and patients) that provide information to, and maintain, the ANZDATA database. The data reported here have been supplied by ANZDATA. The interpretation and reporting of these data are the responsibility of the authors and in no way should be seen as official policy or interpretation of ANZDATA.
We also would like to thank Dr. Jenny Lalor for her assistance with data analysis.

Funding

No specific funding for this study was provided.

Availability of data and materials

The data that support the findings of this study are available from ANZDATA upon ethics committee approval and approval by the ANZDATA committee.
Data obtained from the ANZDATA registry included the patient unique medical record number (UMRN) to enable matching with laboratory data and obtaining information from patient charts. After obtaining the additional data, the UMRNs were removed from the record and the ANZDATA patient record number was used as the identifier. Patient consent was not obtained for this study as it was impractical to obtain patient consent and because the risk of adverse outcomes from the study is very low. Approval for the study was obtained from the Sir Charles Gairdner Hospital Human Research Ethics Committee (approval number 2015–022) and from the Curtin University Human Research Ethics Committee (approval number HR64/2015).
Not applicable.

Competing interests

The authors 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
3.
6.
Zurück zum Zitat Courivaud C, Bamoulid J, Chalopin JM, Gaiffe E, Tiberghien P, Saas P, Ducloux D. Cytomegalovirus exposure and cardiovascular disease in kidney transplant recipients. J Infect Dis. 2013;207:1569–75. doi:10.1093/infdis/jit064.CrossRefPubMed Courivaud C, Bamoulid J, Chalopin JM, Gaiffe E, Tiberghien P, Saas P, Ducloux D. Cytomegalovirus exposure and cardiovascular disease in kidney transplant recipients. J Infect Dis. 2013;207:1569–75. doi:10.​1093/​infdis/​jit064.CrossRefPubMed
8.
Zurück zum Zitat Hodson EM, Ladhani M, Webster AC, Strippoli GF, Craig JC. Antiviral medications for preventing cytomegalovirus disease in solid organ transplant recipients. Cochrane Database Syst Rev. 2013:CD003774. doi:10.1002/14651858.CD003774.pub4. Hodson EM, Ladhani M, Webster AC, Strippoli GF, Craig JC. Antiviral medications for preventing cytomegalovirus disease in solid organ transplant recipients. Cochrane Database Syst Rev. 2013:CD003774. doi:10.​1002/​14651858.​CD003774.​pub4.
9.
Zurück zum Zitat Humar A, Lebranchu Y, Vincenti F, Blumberg EA, Punch JD, Limaye AP, Abramowicz D, Jardine AG, Voulgari AT, Ives J, et al. The efficacy and safety of 200 days valganciclovir cytomegalovirus prophylaxis in high-risk kidney transplant recipients. Am J Transplant. 2010;10:1228–37. doi:10.1111/j.1600-6143.2010.03074.x.CrossRefPubMed Humar A, Lebranchu Y, Vincenti F, Blumberg EA, Punch JD, Limaye AP, Abramowicz D, Jardine AG, Voulgari AT, Ives J, et al. The efficacy and safety of 200 days valganciclovir cytomegalovirus prophylaxis in high-risk kidney transplant recipients. Am J Transplant. 2010;10:1228–37. doi:10.​1111/​j.​1600-6143.​2010.​03074.​x.CrossRefPubMed
10.
Zurück zum Zitat Razonable RR, Rivero A, Rodriguez A, Wilson J, Daniels J, Jenkins G, Larson T, Hellinger WC, Spivey JR, Paya CV. Allograft rejection predicts the occurrence of late-onset cytomegalovirus (CMV) disease among CMV-mismatched solid organ transplant patients receiving prophylaxis with oral ganciclovir. J Infect Dis. 2001;184:1461–4.CrossRefPubMed Razonable RR, Rivero A, Rodriguez A, Wilson J, Daniels J, Jenkins G, Larson T, Hellinger WC, Spivey JR, Paya CV. Allograft rejection predicts the occurrence of late-onset cytomegalovirus (CMV) disease among CMV-mismatched solid organ transplant patients receiving prophylaxis with oral ganciclovir. J Infect Dis. 2001;184:1461–4.CrossRefPubMed
11.
Zurück zum Zitat Harvala H, Stewart C, Muller K, Burns S, Marson L, MacGilchrist A, Johannessen I. High risk of cytomegalovirus infection following solid organ transplantation despite prophylactic therapy. J Med Virol. 2013;85:893–8. doi:10.1002/jmv.23539.CrossRefPubMed Harvala H, Stewart C, Muller K, Burns S, Marson L, MacGilchrist A, Johannessen I. High risk of cytomegalovirus infection following solid organ transplantation despite prophylactic therapy. J Med Virol. 2013;85:893–8. doi:10.​1002/​jmv.​23539.CrossRefPubMed
12.
Zurück zum Zitat San-Juan R, De Dios B, Garcia-Reyne A, Fernandez-Ruiz M, Lumbreras C, Lopez-Medrano F, Morales JM, Hernando S, Folgueira D, Jimenez C, Aguado JM. Limited impact of cytomegalovirus infection in the long-term outcome of renal and liver transplant. J Clin Virol. 2013;56:316–22. doi:10.1016/j.jcv.2012.12.010.CrossRefPubMed San-Juan R, De Dios B, Garcia-Reyne A, Fernandez-Ruiz M, Lumbreras C, Lopez-Medrano F, Morales JM, Hernando S, Folgueira D, Jimenez C, Aguado JM. Limited impact of cytomegalovirus infection in the long-term outcome of renal and liver transplant. J Clin Virol. 2013;56:316–22. doi:10.​1016/​j.​jcv.​2012.​12.​010.CrossRefPubMed
14.
Zurück zum Zitat Pang XL, Fox JD, Fenton JM, Miller GG, Caliendo AM, Preiksaitis JK. American Society of Transplantation infectious diseases Community of P, Canadian Society of T: Interlaboratory comparison of cytomegalovirus viral load assays. Am J Transplant. 2009;9:258–68. doi:10.1111/j.1600-6143.2008.02513.x.CrossRefPubMed Pang XL, Fox JD, Fenton JM, Miller GG, Caliendo AM, Preiksaitis JK. American Society of Transplantation infectious diseases Community of P, Canadian Society of T: Interlaboratory comparison of cytomegalovirus viral load assays. Am J Transplant. 2009;9:258–68. doi:10.​1111/​j.​1600-6143.​2008.​02513.​x.CrossRefPubMed
15.
Zurück zum Zitat Veroux M, Grosso G, Corona D, Mistretta A, Giaquinta A, Giuffrida G, Sinagra N, Veroux P. Age is an important predictor of kidney transplantation outcome. Nephrol Dial Transplant. 2012;27:1663–71. doi:10.1093/ndt/gfr524.CrossRefPubMed Veroux M, Grosso G, Corona D, Mistretta A, Giaquinta A, Giuffrida G, Sinagra N, Veroux P. Age is an important predictor of kidney transplantation outcome. Nephrol Dial Transplant. 2012;27:1663–71. doi:10.​1093/​ndt/​gfr524.CrossRefPubMed
16.
Zurück zum Zitat Meier-Kriesche HU, Ojo AO, Cibrik DM, Hanson JA, Leichtman AB, Magee JC, Port FK, Kaplan B. Relationship of recipient age and development of chronic allograft failure. Transplantation. 2000;70:306–10.CrossRefPubMed Meier-Kriesche HU, Ojo AO, Cibrik DM, Hanson JA, Leichtman AB, Magee JC, Port FK, Kaplan B. Relationship of recipient age and development of chronic allograft failure. Transplantation. 2000;70:306–10.CrossRefPubMed
17.
Zurück zum Zitat Frei U, Noeldeke J, Machold-Fabrizii V, Arbogast H, Margreiter R, Fricke L, Voiculescu A, Kliem V, Ebel H, Albert U, et al. Prospective age-matching in elderly kidney transplant recipients--a 5-year analysis of the Eurotransplant senior program. Am J Transplant. 2008;8:50–7. doi:10.1111/j.1600-6143.2007.02014.x.PubMed Frei U, Noeldeke J, Machold-Fabrizii V, Arbogast H, Margreiter R, Fricke L, Voiculescu A, Kliem V, Ebel H, Albert U, et al. Prospective age-matching in elderly kidney transplant recipients--a 5-year analysis of the Eurotransplant senior program. Am J Transplant. 2008;8:50–7. doi:10.​1111/​j.​1600-6143.​2007.​02014.​x.PubMed
24.
Zurück zum Zitat Lim WH, Eris J, Kanellis J, Pussell B, Wiid Z, Witcombe D, Russ GR. A systematic review of conversion from calcineurin inhibitor to mammalian target of rapamycin inhibitors for maintenance immunosuppression in kidney transplant recipients. Am J Transplant. 2014;14:2106–19. doi:10.1111/ajt.12795.CrossRefPubMed Lim WH, Eris J, Kanellis J, Pussell B, Wiid Z, Witcombe D, Russ GR. A systematic review of conversion from calcineurin inhibitor to mammalian target of rapamycin inhibitors for maintenance immunosuppression in kidney transplant recipients. Am J Transplant. 2014;14:2106–19. doi:10.​1111/​ajt.​12795.CrossRefPubMed
25.
Zurück zum Zitat Brennan DC, Legendre C, Patel D, Mange K, Wiland A, McCague K, Shihab FS. Cytomegalovirus incidence between Everolimus versus Mycophenolate in de novo renal transplants: pooled analysis of three clinical trials. Am J Transplant. 2011;11:2453–62.CrossRefPubMed Brennan DC, Legendre C, Patel D, Mange K, Wiland A, McCague K, Shihab FS. Cytomegalovirus incidence between Everolimus versus Mycophenolate in de novo renal transplants: pooled analysis of three clinical trials. Am J Transplant. 2011;11:2453–62.CrossRefPubMed
27.
Zurück zum Zitat Martin-Gandul C, Perez-Romero P, Sanchez M, Bernal G, Suarez G, Sobrino M, Merino L, Cisneros JM, Cordero E. Spanish network for research in infectious D: determination, validation and standardization of a CMV DNA cut-off value in plasma for preemptive treatment of CMV infection in solid organ transplant recipients at lower risk for CMV infection. J Clin Virol. 2013;56:13–8. doi:10.1016/j.jcv.2012.09.017.CrossRefPubMed Martin-Gandul C, Perez-Romero P, Sanchez M, Bernal G, Suarez G, Sobrino M, Merino L, Cisneros JM, Cordero E. Spanish network for research in infectious D: determination, validation and standardization of a CMV DNA cut-off value in plasma for preemptive treatment of CMV infection in solid organ transplant recipients at lower risk for CMV infection. J Clin Virol. 2013;56:13–8. doi:10.​1016/​j.​jcv.​2012.​09.​017.CrossRefPubMed
28.
Zurück zum Zitat Razonable RR, Humar A. Cytomegalovirus in solid organ transplantation. Am J Transplant. 2013;13:93–106.CrossRefPubMed Razonable RR, Humar A. Cytomegalovirus in solid organ transplantation. Am J Transplant. 2013;13:93–106.CrossRefPubMed
29.
Zurück zum Zitat Torre-Cisneros J, Aguado JM, Caston JJ, Almenar L, Alonso A, Cantisan S, Carratala J, Cervera C, Cordero E, Farinas MC, et al. Management of cytomegalovirus infection in solid organ transplant recipients: SET/GESITRA-SEIMC/REIPI recommendations. Transplant Rev (Orlando). 2016;30:119–43. doi:10.1016/j.trre.2016.04.001.CrossRef Torre-Cisneros J, Aguado JM, Caston JJ, Almenar L, Alonso A, Cantisan S, Carratala J, Cervera C, Cordero E, Farinas MC, et al. Management of cytomegalovirus infection in solid organ transplant recipients: SET/GESITRA-SEIMC/REIPI recommendations. Transplant Rev (Orlando). 2016;30:119–43. doi:10.​1016/​j.​trre.​2016.​04.​001.CrossRef
31.
Zurück zum Zitat Arthurs SK, Eid AJ, Pedersen RA, Kremers WK, Cosio FG, Patel R, Razonable RR. Delayed-onset primary cytomegalovirus disease and the risk of allograft failure and mortality after kidney transplantation. Clin Infect Dis. 2008;46:840–6. doi:10.1086/528718.CrossRefPubMed Arthurs SK, Eid AJ, Pedersen RA, Kremers WK, Cosio FG, Patel R, Razonable RR. Delayed-onset primary cytomegalovirus disease and the risk of allograft failure and mortality after kidney transplantation. Clin Infect Dis. 2008;46:840–6. doi:10.​1086/​528718.CrossRefPubMed
Metadaten
Titel
Cytomegalovirus viraemia and mortality in renal transplant recipients in the era of antiviral prophylaxis. Lessons from the western Australian experience
verfasst von
Linda A. Selvey
Wai H. Lim
Peter Boan
Ramyasuda Swaminathan
Claudia Slimings
Amy E. Harrison
Aron Chakera
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
BMC Infectious Diseases / Ausgabe 1/2017
Elektronische ISSN: 1471-2334
DOI
https://doi.org/10.1186/s12879-017-2599-y

Weitere Artikel der Ausgabe 1/2017

BMC Infectious Diseases 1/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

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