You are on page 1of 8

The Journal of Infectious Diseases

MAJOR ARTICLE

Glomerular Parietal Epithelial Cells Infection Is Associated


With Poor Graft Outcome in Kidney Transplant Recipients
With BK Polyomavirus–Associated Nephropathy

Downloaded from https://academic.oup.com/jid/article-abstract/219/12/1879/5288592 by F. Hoffmann-La Roche Ltd user on 27 November 2019


Xu-Tao Chen,1,a Shi-Cong Yang,2,a Wen-Fang Chen,2 Jun Li,1 Su-Xiong Deng,1 Jiang Qiu,1 Ji-Guang Fei,1 Rong-Hai Deng,1 Yan-Yang Chen,2 Pei-Song Chen,3
Yang Huang,1 Chang-Xi Wang,1,b and Gang Huang1,b
1
Organ Transplant Center, 2Department of Pathology, and 3Clinical Laboratory Department, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China

Background.  The purpose of this study was to investigate the effect of BK polyomavirus (BKPyV infection of glomerular parietal
epithelial cells (GPECs) on graft outcome in kidney transplant recipients with BKPyV-associated nephropathy (BKPyVAN).
Methods.  A total of 152 kidney transplant recipients with BKPyVAN were divided into 31 with (GPEC-positive group) and 121
without (GPEC-negative group) BKPyV-infected GPECs. Clinicopathological characteristics and allograft survival were compared
between the groups.
Results.  The GPEC-positive group had more patients with advanced-stage BKPyVAN than the GPEC-negative group (P < .001).
At the last follow-up, the GPEC-positive group had a significantly higher serum creatinine level than the GPEC-negative group. The
graft loss rate in the GPEC-positive group was higher than that in the GPEC-negative group (32.3% vs 12.4%; P = .008). Kaplan-
Meier analysis showed that the graft survival rate in the GPEC-positive group was lower than that in the GPEC-negative group (log-
rank test, P = .004). Multivariate Cox regression analysis demonstrated that BKPyV infection of GPECs was an independent risk
factor for graft survival (hazard ratio, 3.54; 95% confidence interval, 1.43–8.76; P = .006).
Conclusions.  GPEC infection in patients with BKPyVAN indicates more-severe pathological damage and a rapid decline in
renal function. BKPyV infection of GPECs is an independent risk factor for allograft loss.
Keywords.  BK polyomavirus; renal transplantation; glomerular infection; pathology; graft survival.

BK polyomavirus (BKPyV)–associated nephropathy tubular involvement, we have noticed that BKPyV infection
(BKPyVAN) is a severe complication of renal transplantation, can be occasionally observed in the Bowman space in renal bi-
affecting 1%–10% of kidney transplant recipients in the first opsy specimens from patients with BKPyVAN. Nevertheless,
2 years after transplantation [1]. Immunosuppression following glomerular BKPyV infection received minimal attention, and
kidney transplantation has been shown to increase the risk of scant literature has described this phenomenon [9–11]. Celik
BKPyV reactivation in recipients [2–4]. At present, there is no et al reviewed 124 biopsy specimens from 83 kidney transplant
effective antiviral treatment for BKPyV infection, and recipients recipients and identified 36 specimens with BKPyV infection
with BKPyVAN have an elevated risk of graft loss [1]. It has of the Bowman space [11]. However, the effect of glomerular
been reported that renal graft failure occurs in 10%–80% of BKPyV infection on graft outcome is unknown.
recipients within 2–3 years after diagnosis of BKPyVAN [1, 5]. There are 3 histological classifications of BKPyVAN, in-
The typical pathological features of BKPyVAN include cluding the original schema reported by the University of
tubulointerstitial lesions, viral cytopathic damage to tubular epi- Maryland [12] and subsequently modified by American
thelial cells, and positive results of immunohistochemical (IHC) Society of Transplantation (AST) [8], as well as another system
nuclear staining for SV40 large T antigen [6–8]. In addition to proposed by the Banff Working Group [7]. These classifications
focus on several pathological features of BKPyVAN, such as

cytopathic effects, tubulitis, interstitial inflammation, intersti-
Received 25 November 2018; editorial decision 4 January 2019; accepted 9 January 2019; tial fibrosis, and tubular atrophy, which have been shown to
published online January 12, 2019.
a
X.-T. C. and S.-C. Y. contributed equally to this work. be associated with graft loss [6, 7, 13]. However, none of these
b
G. H. and C.-X. W. contributed equally to this work. histological classifications suggests a clinical significance of glo-
Correspondence: G. Huang, MD, PhD, #58 Zhongshan Rd 2, Guangzhou, Guangdong Province,
China, 510080 (huanggang_791021@163.com). merular infection in patients with BKPyVAN. Thus, the pur-
The Journal of Infectious Diseases®  2019;219:1879–86 pose of this study was to examine the effect of BKPyV infection
© The Author(s) 2019. Published by Oxford University Press for the Infectious Diseases Society
of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.
of glomerular parietal epithelial cells (GPECs) on graft outcome
DOI: 10.1093/infdis/jiz022 in kidney transplant recipients with BKPyVAN.

Glomerular infection in BKPyVAN  •  jid 2019:219 (15 June) • 1879


METHODS viral inclusions (decoy cells), which were counted as the number
Study Design and Patients
per 10 high-power fields [14].
The medical records of 897 kidney transplant recipients who un-
Virological Studies
derwent kidney allograft biopsies at our hospital between 2007
Urine and plasma BKPyV loads were quantitatively measured
and 2017 were retrospectively reviewed. A flow diagram of pa-
by quantitative polymerase chain reaction (PCR) analysis (MJ
tient inclusion is shown in Figure 1. A total of 159 patients who
Research, Waltham, MA). Specimen collection and processing,
had at least 1 biopsy specimen that was positive for anti–SV40
PCR primers, the TaqMan probe (targeting the BKPyV VP1
large T antigen on immunohistochemical (IHC) staining were
gene), the plasmid standard containing the targeted BKPyV

Downloaded from https://academic.oup.com/jid/article-abstract/219/12/1879/5288592 by F. Hoffmann-La Roche Ltd user on 27 November 2019


identified in the medical records. Seven patients were excluded:
VP1 gene, amplification protocols, PCR precautions, and
3 patient was excluded because of an insufficient number of
quality assurance were performed as previously described [15].
glomeruli (ie, <7) in the biopsy specimen; 1, because of concur-
Urine and plasma BKPyV loads are presented as BKPyV ge-
rent myeloma nephropathy; and 3, because of positivity for JC
nome copies per milliliter. The limit of quantitation was 1000
virus viremia. The remaining 152 patients were divided into 31
copies/mL.
with (GPEC-positive group) and 121 without (GPEC-negative
group) BKPyV-infected GPECs, based on the presence or ab- Diagnosis of BKPyVAN
sence of anti-SV40 large T antigen on IHC staining. Specimens were stained with hematoxylin and eosin, periodic
This study adhered to the tenets of the Declaration of Helsinki acid Schiff, and Masson silver trichrome stains for light micros-
and was approved by the ethics committee and research board copy. The diagnosis of polyomavirus-associated nephropathy
of our institution. Written informed consent was obtained from (PyVAN) was established by the presence of interstitial inflam-
each patient. mation, tubular atrophy, and interstitial fibrosis and the extent
of viral cytopathic changes in the tubular epithelial cells, and it
Data Collection
was confirmed by positive results of IHC nuclear staining with
Patient demographic and clinicopathological data were col-
anti-SV40 large T antigen monoclonal antibody, as previously
lected and compared between the 2 groups. Severe pneumonia
described [16]. The histological features of PyVAN were classi-
was defined as the requirement of any mechanical ventila-
fied using the AST schema, and PyVAN was classified as stage A,
tion. Delayed graft function was defined as the requirement
B, and C, based on the guidelines published by Hirsch et al [8].
of any dialysis treatment within the first week after trans-
Histological evaluation of the viral load was semiquantitative,
plantation. The end point of the analysis was the date of pa-
with findings reported as the percentage of tubules positive for
tient death or graft loss (ie, the date of dialysis resumption or
polyomavirus, using a 4-tier system (<10%, 10%–25%, 25%–
retransplantation) or the date of the most recent clinical data
50%, and >50%) [15]. The diagnosis of BKPyVAN was con-
(until December 2017).
firmed on the basis of positive BK viruria and/or BK viremia,
Urine Cytology in addition to positive results of immunostaining for SV40
Urinary cytological smears were stained by the Papanicolaou large T antigen. Particular attention was paid to the presence
method and evaluated for the presence of cells with intranuclear of anti-SV40 large T antigen staining in the Bowman space.

Kidney transplantation n = 897


(allograft biopsy n = 1200)

Inclusion criteria
• Positive SV40 large T antigen

PyVANa n = 159

Exclusion criteria
• Concurrent myeloma
nephropathy n = 1
• JC polyomavirus
infection n = 3
GPEC positive n = 31 GPEC negative n = 121 • Glomeruli <7 n = 3

Figure 1.  Flow of patients through the study. GPEC, glomerular parietal epithelial cell; PyVAN, polyomavirus-associated nephropathy. aBetween 2007 and 2016.

1880 • jid 2019:219 (15 June) • Chen et al


GPEC-positive was defined as at least 1 glomerulus positive for significance. All analyses were performed by using IBM SPSS,
anti-SV40 large T antigen. All slides were reviewed by 2 inde- version 20 (SPSS Statistics V20, IBM, Somers, NY).
pendent pathologists, and histological scores were evaluated
using the Banff criteria [7, 17, 18]. RESULTS

Patient Characteristics
Electron Microscopy
Patient characteristics are summarized in Table 1. The base-
A Philips CM10 electron microscope (Philips, Eindhoven, the
line serum creatinine level in the GPEC-negative group
Netherlands) was used to observe viral particles, especially in
was significantly higher than in the GPEC-positive group
tubular epithelial cells and the Bowman space [16]. Cells with

Downloaded from https://academic.oup.com/jid/article-abstract/219/12/1879/5288592 by F. Hoffmann-La Roche Ltd user on 27 November 2019


(P = .004, Table 1). However, there was no significant differ-
viral particles (diameter, approximately 45  nm) in the cyto-
ence in other characteristics between the GPEC-positive and
plasm or nucleus were considered to be infected.
GPEC-negative groups, including sex, age, etiology of end-
Modification of Immunosuppression and Follow-up
stage renal disease, duration and type of dialysis, immunosup-
For patients with BKPyVAN, modification of immunosuppres- pressive regimen, donor type, severe pneumonia, and delayed
sion included a reduction in the dosage of calcineurin inhibitor graft function.
and a switch from tacrolimus treatment to cyclosporine A  or
Time of BKPyV Infection of GPECs
rapamycin treatment. For patients with severe infections, the
The time of BKPyV infection of GPECs in the GPEC-positive
dosage of mycophenolic acid and/or calcineurin inhibitor was
group is shown in Supplementary Figure 1. A significant pro-
reduced, or the medication(s) was discontinued. The adminis-
portion of patients with BKPyV-infected GPECs were identified
tration of oral glucocorticoids was not changed. Acute cellular
within 24 months after transplantation, with others developing
rejection was treated with pulse methylprednisolone with or
BKPyV-infected GPECs up to 7 years after transplantation.
without polyclonal antibody. Antibody-mediated rejection was
treated with pulse methylprednisolone, intravenous immuno- Comparison of BKPyV Load Between Groups
globulin, rituximab, and/or plasma exchange. At the initial diagnosis of BKPyVAN, the number of decoy cells
All patients were regularly followed up in the outpatient and BKPyV loads in the urine and plasma were not significantly
clinic, and serum creatinine level, calcineurin inhibitor trough different between the GPEC-positive and GPEC-negative
level, and BKPyV load were monitored. Follow-up renal biopsy groups (all, P  >  .05, Supplementary Table 1). Treatments for
was performed if the serum creatinine level was continuously BKPyVAN were not significantly different between the 2 groups
elevated or after treatment of BKPyVAN for 6–12 months. (P = .491; Supplementary Table 2).
During follow-up, there was no significant difference in
Statistical Analysis
the clearance of BKPyV DNA from blood (83.9% vs 92.6%;
Continuous data are expressed as means ± standard deviations
P = .163) and urine (19.4% vs 22.3%; P = .721; Supplementary
and were compared by the Student independent t test. If nor-
Table 2) between the 2 groups.
mality was not assumed, the Mann-Whitney U test was use to
compare data between groups. Categorical data are presented as Comparison of Graft Function Between Groups
numbers and percentages were and compared by the Pearson χ2 The vast majority of patients in both groups underwent a renal
test or the Fisher exact test (if an expected value was ≤5). Two- biopsy because of an elevated serum creatinine level (>30%
factor mixed-design analysis of variance (ANOVA; 1 variable above baseline). The serum creatinine level at initial diagnosis
was time dependent) was used to compare serum creatinine of BKPyVAN was significantly elevated, compared with the level
level at baseline, diagnosis, and at final follow-up. Point-biserial at a stable phase in both groups (P < .05 for both comparisons).
correlation analysis was used to calculate the correlation co- There was no significant difference in serum creatinine level at
efficient between pathological scores and BKPyV infection the initial diagnosis of BKPyVAN between groups (P  =  .352;
in the glomerulus. Survival analysis using the Kaplan-Meier Supplementary Table 1). However, the mean serum creatinine
method and a Cox proportional-hazards regression model was level (±SD) at the last follow-up visit was significantly higher
performed to evaluate the impact of BKPyV infection of GPECs in the GPEC-positive group, compared with that in the GPEC-
on allograft survival. To investigate the association of BKPyV negative group (255.43  ±  150.71 vs 196.60  ±  112.50  μmol/L;
infection of GPECs and other independent variables with al- P = .041; Supplementary Table 2).
lograft survival, univariate and multivariate regression models The median duration of follow-up was 11.2  months in the
were performed, and only variables significant in both univar- GPEC-positive group and 19.6  months in the GPEC-negative
iate and multivariate results were considered associated factors. group. The rate of increase in the serum creatinine level was
Results are reported as estimated hazard ratios (HRs) and 95% greater in the GPEC-positive group than in the GPEC-negative
confidence intervals (CIs). All statistical tests were 2-tailed, group (P < .001 for time effect; P = .003 for time × GPEC effect;
and a P value of <  .05 was considered to indicate statistical 2-factor mixed-design ANOVA).

Glomerular infection in BKPyVAN  •  jid 2019:219 (15 June) • 1881


Table 1.  Demographic and Transplant Characteristics of 152 Patients With BK Polyomavirus (BKPyV)–Associated Nephropathy, by BKPyV Infection Status
in Glomerular Parietal Epithelial Cells (GPECs)

GPEC Negative GPEC Positive Total


Parameter (n = 121) (n = 31) (n = 152) P

Sex .568
 Male 77 (63.64) 18 (58.06) 95 (62.50)
 Female 44 (36.36) 13 (41.94) 57 (37.50)
Age at transplantation, y 38.78 ± 8.88 41.15 ± 9.71 39.27 ± 9.08 .195

Downloaded from https://academic.oup.com/jid/article-abstract/219/12/1879/5288592 by F. Hoffmann-La Roche Ltd user on 27 November 2019


ESRD etiology .500
  Chronic glomerulonephritis 78 (64.46) 23 (74.19) 101 (66.45)
  IgA nephropathy 17 (14.05) 4 (12.90) 21 (13.82)
 Other 26 (21.49) 4 (12.90) 30 (19.74)
Renal replacement therapy .601
  Peritoneal dialysis 29 (23.97) 6 (19.35) 35 (23.03)
 Hemodialysis 83 (68.60) 21 (67.74) 104 (68.42)
  Without dialysis 9 (7.44) 4 (12.90) 13 (8.55)
Immunosuppressive regimen
  Polyclonal antibody 64 (52.89) 18 (58.06) 82 (53.95) .606
  Monoclonal antibody 51 (42.15) 12 (38.71) 63 (41.45) .729
 Tac-MPA 120 (99.17) 31 (100.00) 151 (99.34) 1.000
 CsA-MPA 1 (0.83) 0 (0.00) 1 (0.66) 1.000
Donor type
 Living 22 (18.18) 5 (16.13) 27 (17.76) 1.000
 Nonliving 99 (81.82) 26 (83.87) 125 (82.24) 1.000
Baseline creatinine level at stable phase, μmol/L 129.52 ± 44.66 104.97 ± 25.85 124.51 ± 42.62 .004
Complication
  Delayed graft function 15 (12.40) 4 (12.90) 19 (12.50) 1.000
  Severe pneumonia 14 (11.57) 5 (16.13) 19 (12.50) .544
  Preceding TCMR/AMR 8 (6.61) 2 (6.45) 10 (6.58) 1.000

Data are no. (%) of participants or mean value ± SD.


Abbreviations: AMR, antibody-mediated rejection; CsA, cyclosporine A; ESRD, end-stage renal disease; IgA, immunoglobulin A; MPA, mycophenolic acid; Tac, tacrolimus; TCMR, T cell–
mediated rejection.

More patients developed a sustained increase in serum stage BKPyVAN than the GPEC-negative group (P  <  .001;
creatinine level (>30% greater than that at diagnosis) in the Supplementary Table 3). The incidence of periglomerular fi-
GPEC-positive group than in the GPEC-negative group brosis in the GPEC-positive group was higher than that in
(41.9% vs 18.2%; P = .005; Supplementary Table 2). The abso- the GPEC-negative group (P  =  .045; Supplementary Table 3).
lute value of the increase in serum creatinine level (calculated Positive anti-SV40 large T antigen staining in tubular epithelial
as the level at the last follow-up visit minus the level first re- cells was observed in all biopsy specimens from both groups.
corded) was significantly higher in the GEPC-positive group Notably, the mean Banff scores for the extent of SV40 large
than that in the GEPC-negative group (304.71  ±  329.89 vs T antigen staining, tubulitis, interstitial inflammation, tubular
115.74 ± 217.63 μmol/L; P < .001). These results suggest that the atrophy, and interstitial fibrosis were all significantly higher in
GPEC-positive group had worse graft function than the GPEC- the GPEC-positive group, compared with the GPEC-negative
negative group. group (P  <  .05 for all comparisons; Supplementary Table 3).
Correlation analysis revealed that positivity in GPECs was sig-
Pathological Findings at Initial Diagnosis of BKPyVAN nificantly positively correlated with the extent of SV40 large T
Representative images of GPEC-positive biopsy specimens antigen staining (r  =  0.52), tubulitis (r  =  0.17), interstitial in-
under light microscopy and electron microscopy are shown in flammation (r = 0.30), tubular atrophy (r = 0.21), and interstitial
Figure 2. Based on AST criteria, in the GPEC-positive group fibrosis (r = 0.28; P < .05 for all comparisons).
there were no patients with stage A  disease, 26 (83.9%) with
stage B disease, and 5 (16.1%) with stage C disease. In the Pathological Findings During Follow-up
GPEC-negative group, there were 11 patients (9.1%) with stage During follow-up, 12 of 31 patients in the GPEC-positive
A disease, 96 (79.3%) with stage B disease, and 14 (11.6%) with group and 61 of 121 patients in the GPEC-negative group un-
stage C disease (Supplementary Table 3). The GPEC-positive derwent repeat biopsies (Supplementary Table 3). The median
group had a higher proportion of patients with advanced time between the initial biopsy and last follow-up biopsy was

1882 • jid 2019:219 (15 June) • Chen et al


group, compared with that in the GPEC-negative group (log-
rank test P  =  .004; Figure 3). The mean time (±SD) from di-
agnosis of BKPyVAN to graft loss was 7.4 ± 8.5 months in the
GPEC-positive group and 17.2  ±  20.5  months in the GPEC-
negative group (P = .179).
Univariate and multivariate Cox regression models were
performed to investigate risk factors for graft loss. As shown in
A B Table 2, BKPyV infection of GPECs was identified as an inde-

Downloaded from https://academic.oup.com/jid/article-abstract/219/12/1879/5288592 by F. Hoffmann-La Roche Ltd user on 27 November 2019


pendent risk factor for graft loss (HR, 3.54; 95% CI, 1.43–8.76;
P = .006). Serum creatinine level at diagnosis (HR, 1.01; 95%
CI, 1.00–1.01; P  <  .001) and tubular atrophy score (HR, 3.13;
95% CI, 1.21–8.12; P = .019) were also independent risk factors
for graft loss.

C D DISCUSSION

In this study, we investigated the effect of GPEC infection on


Figure 2.  Histological features in biopsy specimens from patients with BK graft outcome in kidney transplant recipients with BKPyVAN.
polyomavirus (BKPyV)–associated nephropathy with glomerular parietal epithe-
Of the patients with BKPyVAN, 20.4% developed a GPEC in-
lial cell (GPEC) involvement. A, BKPyV-involved GPECs exhibited an enlarged and
ground-grass–like nucleus, accompanied with prominent intranuclear inclusions fection, most within 24 months after transplantation. At the last
(arrows; hematoxylin and eosin staining; original magnification  ×200). B, follow-up, the GPEC-positive group had a significantly higher
Immunohistochemical staining showed nuclear positivity for SV40 large T antigen serum creatinine level and ratio of sustained increase in serum
(arrows) in GPECs (DAB staining; original magnification ×200). C, Electronic micros-
copy revealed electron-dense structures in the nucleus of GPECs (arrow; original creatinine level. At diagnosis of BKPyVAN, the proportion of
magnification ×1500). D, On high-power microscopy, these structures had a para- patients with advanced-stage BKPyVAN was greater in the
crystalline arrangement, composing of naked and round viral particles that meas- GPEC-positive group. The graft loss rate in the GPEC-positive
ured 45 nm in diameter (original magnification ×50 000).
group was significantly higher than that in the GPEC-negative
group. Kaplan-Meier analysis showed that the graft survival
rate in the GPEC-positive group was significantly lower than
14.35 months (range, 1.63–39.47 months) in the GPEC-positive
that in the GPEC-negative group. Multivariate Cox regression
group and 16.45  months (range, 0.30–69.23  months) in the
analysis demonstrated that GPEC infection was an independent
GPEC-negative group.
risk factor for graft loss. Taken together, these results suggested
The proportion of patients who developed global
glomerulosclerosis was similar between the 2 groups (2.5% vs
2.4%). In the GPEC-positive group, compared with findings for the
initial biopsy specimen, SV40 large T antigen staining, tubulitis, 1.0 GPEC
GPEC negative
and interstitial inflammation were decreased but tubular atrophy GPEC positive

and interstitial fibrosis were increased on the last biopsy spec- GPEC negative censored
GPEC positive censored
0.8
imen (Supplementary Table 3). During follow-up, the degrees of
Cumulative survival (proportion)

interstitial fibrosis (P = .043) and tubular atrophy (P = .040) were


significantly higher in the GPEC-positive group, compared with 0.6
findings in the GPEC-negative group (Supplementary Table 3).
Pathological changes in biopsy specimens from the first to the last
biopsy are summarized in Supplementary Table 4. 0.4

Association Between GPEC Positivity and Graft Survival


0.2
During follow-up, graft loss in the GPEC-positive group was
significantly greater than that in the GPEC-negative group
(32.3% vs 12.4%; P = .008; Supplementary Table 2). One patient 0
with graft function in the GPEC-positive group died because of 0 25.0 50.0 75.0 100.0 125.0
hepatic failure. The 5-year cumulative graft survival rate after Time after initial biopsy (months)

transplantation was 66.7% in the GPEC-positive group and


Figure 3.  Graft survivals among patients with (glomerular parietal epithelial cell
100.0% in the GPEC-negative group (P = .004). Kaplan-Meier
[GPEC]–positive group) and those without (GPEC-negative group) BK polyomavirus
analysis showed that the death-censored graft survival rate (BKPyV)–infected GPECs. Kaplan-Meier curves show death-censored allograft sur-
after initial biopsy was significantly lower in the GPEC-positive vival times after initial biopsy in both groups. Log-rank test, P = .004.

Glomerular infection in BKPyVAN  •  jid 2019:219 (15 June) • 1883


Table 2.  Risk Factors for Graft Loss, According to Cox Regression Analysis

Univariate Multivariate

Parameter HR (95% CI) P HR (95% CI) P

BKPyV infection status in GPECs


 Negative Reference Reference
 Positive 3.75 (1.66–8.47) .002 3.54 (1.43–8.76) .006
Donor type
 Nonliving Reference …

Downloaded from https://academic.oup.com/jid/article-abstract/219/12/1879/5288592 by F. Hoffmann-La Roche Ltd user on 27 November 2019


 Living 0.58 (.17–1.94) .374 …
Serum creatinine level at diagnosis (mmol/L) 1.01 (1.005–1.011) <.001 1.01 (1.00–1.01) <.001
BKPyV viremia level at diagnosis (copies/mL) 1.00 (1.00–1.00) .946 …
Clearance of BKPyV DNA in blood
 No Reference Reference
 Yes 0.27 (.09–.80) .018 0.32 (.09–1.09) .068
Delayed graft function
 No Reference …
 Yes 2.69 (.98–7.37) .055 …
Preceding rejection episodes
 No Reference …
 Yes 1.36 (.56–3.31) .499 …
Rejection episodes after BKPyVAN .279
 No Reference …
  T cell–mediated rejection 4.31 (1.00–18.55) .050 …
  Antibody-mediated rejection 0.00 (.00–.00) .986 …
  Mixed cellular- and antibody-mediated rejection 0.00 (.00–.00) .990 …
Tubulitis score 0.86 (.52–1.43) .572
Tubular atrophy score 2.68 (1.59–4.52) <.001 3.13 (1.21–8.12) .019
Interstitial inflammation score 2.02 (1.25–3.26) .004 0.89 (.45–1.78) .742
Interstitial fibrosis score 2.10 (1.33–3.32) .001 0.53 (.22–1.23) .139

Abbreviations: BKPyV, BK polyomavirus; BKPyVAN, BK polyomavirus–associated nephropathy; CI, confidence interval; GPEC, glomerular parietal epithelial cell; HR, hazard ratio.

that glomerular BKPyV infection is associated with worse graft demonstrating the effectiveness of these treatment regimens. In
function, more-advanced BKPyVAN stage, and a higher rate of the GPEC-positive group, BKPyV replication also presented a
graft loss in kidney transplant recipients with BKPyVAN. To declining trend. In addition, clearance of BK viremia and BK
the best of our knowledge, this is the first study reporting the viruria occurred in 83.9% and 19.4% of patients, respectively.
prognostic value of GPEC infection on graft outcome in kidney These results suggest that, although GPEC positivity represents
transplant recipients with BKPyVAN. a more severe BKPyV infection, patients with BKPyV-infected
Nankivell et al reported that a high level of viremia was asso- GPECs still respond well to treatment.
ciated with a heavy tissue viral load [19]. Thus, we had assumed Celik et al reported that BKPyV infection in the glomerular
that the GPEC-positive group, probably having a heavier tissue epithelium was usually found in biopsy specimens with a high
viral load, might display a higher plasma BKPyV load than that viral load in the tubular epithelium [11]. In line with this obser-
in the GPEC-negative group, but this turned out to be irrelevant. vation, our pathological findings showed that BKPyV infection
On the other hand, hepatitis B virus (HBV) gene mutations were of GPECs was accompanied by more-severe tubulointerstitial
reported to cause glomerular involvement and HBV-associated inflammation, tubular atrophy, interstitial fibrosis, and a greater
glomerulonephritis [20]. Therefore, we thought it plausible that extent of SV40 large T antigen staining in the tubular epithe-
specific variant strains of BKPyV might have greater pathogenic lium, indicating that BKPyV-infected GPECs are associated
potential and viral invasiveness, resulting in GPEC infection, with more-severe pathological damage. Multivariate Cox re-
which might need further investigation. gression analysis showed that tubular atrophy score was an in-
Reducing the immunosuppression intensity is the primary dependent risk factor for graft loss. Study has shown that, in
treatment for BKPyVAN [5]. In this study, changes in the im- severely BKPyV-infected tubules, necrosis of tubular cells and
munosuppressive regimen, including reducing the dosage or tubular basement membrane denudation can be observed [13,
changing the immunosuppressant, were used in the treatment 21]. Assessment of follow-up biopsy specimens in this study
of BKPyVAN. Clearance of BKPyV DNA in the blood was showed that the degree of SV40 large T antigen staining and
observed in 92.6% of patients in the GPEC-negative group, tubulitis in the GPEC-positive group was reduced as compared

1884 • jid 2019:219 (15 June) • Chen et al


to that in initial biopsy specimens and was correlated with Notes
clearance of BK viruria. This improvement may be attributed Acknowledgments.  We thank all of the nurses at the
to immunological reconstitution after reducing the immuno- Department of Organ Transplantation, the First Affiliated
suppressant dosage. However, the extent of tubular atrophy Hospital, Sun Yat-sen University, for their collaboration and
remained significantly higher in the GPEC-positive group dedication to the patients.
during follow-up, suggesting irreversible pathological damage. Financial support.  This work was supported by the National
Our previous study demonstrated that an elevated serum Natural Science Foundation of China (grant 81770749), the
creatinine level at diagnosis was associated with graft func- Natural Science Foundation of Guangdong Province (grant

Downloaded from https://academic.oup.com/jid/article-abstract/219/12/1879/5288592 by F. Hoffmann-La Roche Ltd user on 27 November 2019


tion decline 12  months after diagnosis and with long-term 2017A030313710), and the Fundamental Research Funds for
graft loss in patients with BKPyVAN [16]. In the current the Central Universities (17ykpy29).
study, the mean serum creatinine level at diagnosis was Potential conflicts of interest.  All authors: No reported
219.1 μmol/L in the GPEC-positive group and 198.6 μmol/L conflicts. All authors have submitted the ICMJE Form for
in the GPEC-negative group, both of which were signif- Disclosure of Potential Conflicts of Interest. Conflicts that the
icantly higher than the baseline level. The majority of our editors consider relevant to the content of the manuscript have
patients (92.0% in the GPEC-positive group and 90.2% in the been disclosed.
GPEC-negative group) had an increased serum creatinine
level at diagnosis, suggesting that most patients had severe References

pathological damage. In addition, the increasing rate of in- 1. Sawinski D, Goral S. BK virus infection: an update on di-
crease in the serum creatinine level was higher in the GPEC- agnosis and treatment. Nephrol Dial Transplant 2015;
positive group, compared with that in the GPEC-negative 30:209–17.
group, during follow-up, indicating more-severe BKPyV 2. Schachtner  T, Babel  N, Reinke  P. Different risk factor
infection and pathological damage in the GPEC-positive profiles distinguish early-onset from late-onset BKV-
group. Multivariate Cox regression analysis confirmed that replication. Transpl Int 2015; 28:1081–91.
the serum creatinine level at diagnosis was an independent 3. Borni-Duval C, Caillard S, Olagne J, et al. Risk factors for BK
risk factor for graft loss. In this study, the 5-year cumula- virus infection in the era of therapeutic drug monitoring.
tive allograft survival rate was 66.7% in the GPEC-positive Transplantation 2013; 95:1498–505.
group and 100% in the GPEC-negative group. The 2-year al- 4. Belliere  J, Kamar  N, Mengelle  C, et  al. Pilot conversion
lograft failure rate for all patients was 30.0%, which was the trial from mycophenolic acid to everolimus in ABO-
same as that reported by the Banff Working Group [7] but incompatible kidney-transplant recipients with BK viruria
higher than that (15.4%) reported by Drachenberg et al [13]. and/or viremia. Transpl Int 2016; 29:315–22.
The GPEC-positive group had a significantly lower death- 5. Barth  H, Solis  M, Lepiller  Q, et  al. 45  years after the dis-
censored graft survival rate, and BKPyV infection of GPECs covery of human polyomaviruses BK and JC: time to speed
was identified as a risk factor for graft loss. These results up the understanding of associated diseases and treatment
strongly suggest that GPEC infection by BKPyV predicts approaches. Crit Rev Microbiol 2017; 43:178–95.
worse graft outcome. 6. Masutani  K, Shapiro  R, Basu  A, Tan  H, Wijkstrom  M,
There are some limitations of this study. This was a retrospec- Randhawa  P. The Banff 2009 working proposal for
tive study, and complete pathological data during follow-up polyomavirus nephropathy: a critical evaluation of its utility
were not available for all patients. as a determinant of clinical outcome. Am J Transplant 2012;
In summary, this study found that the presence of BKPyV- 12:907–18.
infected GPECs in renal biopsy specimens from kidney 7. Nickeleit V, Singh HK, Randhawa P, et al.; Banff Working
transplant recipients with BKPyVAN indicates more-severe Group on Polyomavirus Nephropathy. The Banff
pathological damage and is associated with a rapid decline in Working Group classification of definitive polyomavirus
renal function. Furthermore, GPEC infection is an independent nephropathy: morphologic definitions and clinical
risk factor for graft loss. correlations. J Am Soc Nephrol 2018; 29:680–93.
8. Hirsch  HH, Randhawa  P; AST Infectious Diseases
Supplementary Data Community of Practice. BK polyomavirus in solid
Supplementary materials are available at The Journal of Infectious organ transplantation. Am J Transplant 2013; 13 Suppl
Diseases online. Consisting of data provided by the authors 4:179–88.
to benefit the reader, the posted materials are not copyedited 9. Meehan SM, Kraus MD, Kadambi PV, Chang A. Nephron
and are the sole responsibility of the authors, so questions or segment localization of polyoma virus large T antigen in
comments should be addressed to the corresponding author. renal allografts. Hum Pathol 2006; 37:1400–6.

Glomerular infection in BKPyVAN  •  jid 2019:219 (15 June) • 1885


10. Sekulic M, Crary GS, Herrera Hernandez LP. BK polyomavirus 16. Huang G, Wu LW, Yang SC, et al. Factors influencing graft
tubulointerstitial nephritis with urothelial hyperplasia in a outcomes following diagnosis of polyomavirus -associated
kidney transplant. Am J Kidney Dis 2016; 68:307–11. nephropathy after renal transplantation. PLoS One 2015;
11. Celik  B, Randhawa  PS. Glomerular changes in BK virus 10:e142460.
nephropathy. Hum Pathol 2004; 35:367–70. 17. Sis B, Mengel M, Haas M, et al. Banff ‘09 meeting report:
12. Drachenberg  RC, Drachenberg  CB, Papadimitriou  JC, antibody mediated graft deterioration and implemen-
et al. Morphological spectrum of polyoma virus disease in tation of Banff working groups. Am J Transplant 2010;
renal allografts: diagnostic accuracy of urine cytology. Am J 10:464–71.

Downloaded from https://academic.oup.com/jid/article-abstract/219/12/1879/5288592 by F. Hoffmann-La Roche Ltd user on 27 November 2019


Transplant 2001; 1:373–81. 18. B Kopp J. Banff classification of polyomavirus nephropathy:
13. Drachenberg  CB, Papadimitriou  JC, Chaudhry  MR, et  al. a new tool for research and clinical practice. J Am Soc
Histological evolution of BK virus-associated nephropathy: Nephrol 2018; 29:354–5.
importance of integrating clinical and pathological findings. 19. Nankivell  BJ, Renthawa  J, Sharma  RN, Kable  K,
Am J Transplant 2017; 17:2078–91. O’Connell  PJ, Chapman  JR. BK virus nephropathy: histo-
14. Nickeleit V, Klimkait T, Binet IF, et al. Testing for polyomavirus logical evolution by sequential pathology. Am J Transplant
type BK DNA in plasma to identify renal-allograft recipients 2017; 17:2065–77.
with viral nephropathy. N Engl J Med 2000; 342:1309–15. 20. Lu H, Zhu H, Zhou J. S gene mutations of HBV in children
15. Huang  G, Wang  CX, Zhang  L, et  al. Monitoring of with HBV-associated glomerulonephritis. Virol J 2012;
polyomavirus BK replication and impact of preemptive im- 9:59.
munosuppression reduction in renal-transplant recipients 21. Menter  T, Mayr  M, Schaub  S, Mihatsch  MJ, Hirsch  HH,
in China: a 5-year single-center analysis. Diagn Microbiol Hopfer H. Pathology of resolving polyomavirus-associated
Infect Dis 2015; 81:21–6. nephropathy. Am J Transplant 2013; 13:1474–83.

1886 • jid 2019:219 (15 June) • Chen et al

You might also like