ArticlePDF Available

Incidence and risk factors of acute kidney injury, and its effect on mortality among Japanese patients receiving immune check point inhibitors: a single-center observational study

Authors:
  • Teine Keijinkai Medical Center

Abstract and Figures

Background Immune checkpoint inhibitors (ICPis) are associated with multi-organ immune-related adverse effects. Here, we examined the incidence rate, recovery rate, and risk factors of acute kidney injury complicated with ICPis (ICPi-AKI) and evaluted the association between ICPi-AKI and mortality in Japanese patients.Methods We analyzed 152 consecutive patients receiving ICPis between 2015 and 2019. A logistic regression analysis was performed to identify risk factors for ICPi-AKI incidence and Cox regression analysis was performed to evaluate the association between ICPi-AKI and mortality.ResultsThe mean patient age was 67 ± 10 years, with the median baseline serum creatinine level of 0.78 mg/dL. Twenty-seven patients (18%) developed ICPi-AKI, and 19 (73%) of them recovered. Pembrolizumab use and liver diseases were significant risk factors for the ICPi-AKI incidence. During the follow-up, 85 patients (59%) died, 17 patients (63%) with ICPi-AKI and 68 (54%) patients without ICPi-AKI, respectively. The ICPi-AKI incidence was not independently associated with mortality (adjusted hazard ratio, 0.85; 95% confidence intervals, 0.46–1.61).Conclusions Our finding suggest that pembrolizumab use and liver diseases are associated with a higher risk of ICPi-AKI development, but ICPi-AKI did not affect mortality. Future multi-center studies are needed to develop optimal management and prevention strategies for this complication in patients receiving ICPis.
Content may be subject to copyright.
Vol.:(0123456789)
1 3
Clinical and Experimental Nephrology
https://doi.org/10.1007/s10157-020-02008-1
ORIGINAL ARTICLE
Incidence andrisk factors ofacute kidney injury, andits effect
onmortality amongJapanese patients receiving immune check point
inhibitors: asingle‑center observational study
YoshinosukeShimamura1 · ShotaWatanabe1· TakutoMaeda1· KokiAbe1· YayoiOgawa2· HidekiTakizawa1
Received: 16 March 2020 / Accepted: 2 December 2020
© Japanese Society of Nephrology 2021
Abstract
Background Immune checkpoint inhibitors (ICPis) are associated with multi-organ immune-related adverse effects. Here,
we examined the incidence rate, recovery rate, and risk factors of acute kidney injury complicated with ICPis (ICPi-AKI)
and evaluted the association between ICPi-AKI and mortality in Japanese patients.
Methods We analyzed 152 consecutive patients receiving ICPis between 2015 and 2019. A logistic regression analysis
was performed to identify risk factors for ICPi-AKI incidence and Cox regression analysis was performed to evaluate the
association between ICPi-AKI and mortality.
Results The mean patient age was 67 ± 10years, with the median baseline serum creatinine level of 0.78mg/dL. Twenty-
seven patients (18%) developed ICPi-AKI, and 19 (73%) of them recovered. Pembrolizumab use and liver diseases were
significant risk factors for the ICPi-AKI incidence. During the follow-up, 85 patients (59%) died, 17 patients (63%) with
ICPi-AKI and 68 (54%) patients without ICPi-AKI, respectively. The ICPi-AKI incidence was not independently associated
with mortality (adjusted hazard ratio, 0.85; 95% confidence intervals, 0.46–1.61).
Conclusions Our finding suggest that pembrolizumab use and liver diseases are associated with a higher risk of ICPi-AKI
development, but ICPi-AKI did not affect mortality. Future multi-center studies are needed to develop optimal management
and prevention strategies for this complication in patients receiving ICPis.
Keywords Acute kidney injury· Immune checkpoint inhibitor· Mortality· Liver disease· Pembrolizumab
Introduction
The advent of immune checkpoint inhibitors (ICPis) has
revolutionized the care of patients with advanced malig-
nancies [1]. Currently, programmed cell death protein 1
inhibitors, nivolumab and pembrolizumab; programmed cell
death ligand 1 inhibitors, atezolizumab, durvalumab, and
avelumab; and cytotoxic T lymphocyte-associated antigen 4
inhibitor, ipilimumab, are used worldwide. Despite their sub-
stantial clinical effect, ICPis are associated with multi-organ
immune-related adverse effects (irAEs) [1]. Dermatologic,
gastrointestinal, hepatic, and endocrine toxicities are com-
mon irAEs; nephrotoxicity with acute kidney injury (AKI)
primarily due to acute interstitial nephritis has been increas-
ingly reported [26]. Acute kidney injury complicated with
ICPis (ICPi-AKI) is significant as there is evidence that AKI
is associated with a higher risk of subsequent chronic kidney
disease, healthcare utilization rate, and mortality [7, 8]. The
incidence rate of ICPi-AKI is 1.4–30%, and the wide range is
due to substantial heterogeneity across studies [24]. Several
studies reported an association between ICPi-AKI and pro-
ton pump inhibitors (PPIs) [5, 6]. However, the extrapolation
of these results to other populations is limited because these
study populations mostly comprised Caucasians. Although
several ICPi-AKI cases have been reported in Asian patients
[9], to our knowledge, no study has investigated the ICPi-
AKI incidence and its effect on mortality in Asian popula-
tions. Additionally, it has not been validated whether PPI use
is associated with ICPi-AKI in non-Caucasian populations.
Therefore, we conducted a retrospective observational study
* Yoshinosuke Shimamura
yshimamura.tkh@gmail.com
1 Department ofNephrology, Teine Keijinkai Medical Center,
Sapporo, Hokkaido0068555, Japan
2 Hokkaido Renal Pathology Center, Sapporo, Hokkaido,
Japan
Clinical and Experimental Nephrology
1 3
in a Japanese institution to investigate the incidence rate,
recovery rate, and risk factors of ICPi-AKI, and to evaluate
the association between ICPi-AKI and mortality.
Materials andmethods
Study population
This was a single-center retrospective observational study
conducted in Teine Keijinkai Medical Center, which is a
650-bed tertiary referral institution, in Sapporo, Japan. We
retrospectively reviewed medical records of all patients
receiving ICPis and selected 154 consecutive patients who
initiated ICPis for malignancies between March 1, 2015
and October 31, 2019. Baseline laboratory data were avail-
able for all patients. The study was conducted in accord-
ance with the principles of the Declaration of Helsinki and
was approved by the Institutional Review Board of Teine
Keijinkai Medical Center (approval number 2-019096-00).
All study participants provided written informed consent,
and they were presented the right to avoid enrollment in an
opt-out fashion. The study follows the checklist of “Strength-
ening the Reporting of Observational Studies in Epidemiol-
ogy” (STROBE) guidelines [10].
Outcomes anddefinitions
The outcomes were as follows: (1) incidence rate (within
12months of ICPi initiation), recovery rate, and risk fac-
tors of ICPi-AKI and (2) association between ICPi-AKI
and mortality. AKI was defined and staged according to the
Kidney Disease: Improving Global Outcomes criteria [11];
the ICPi-AKI incidence was determined according to the
clinical evaluation of three nephrologists. All AKI cases
were independently reviewed by two board-certified neph-
rologists (Y.S. and K.A.) to determine the most likely etiol-
ogy, and a third board-certified nephrologist (H.T.) resolved
any diagnostic disagreements (n = 2). ICPi-AKI resolution
was defined as recovery of the serum creatinine (sCr) level
to < 0.35mg/dL above the baseline sCr level. Baseline sCr
level was defined as the last available sCr level during the
past 6months since ICPi initiation. The patients were fol-
lowed-up until the study end date (January 31, 2020) or until
the patients were lost to follow-up, had their care withdrawn,
or died.
Covariates
Clinical and pathological data for all participants were
obtained from medical records. The covariates consid-
ered were patient characteristics, including age, sex, and
body mass index [BMI; body weight (kg)/height2 (m2)];
age-adjusted Charlson comorbidity index (ACCI) [12]; the
presence of congestive heart failure, cerebrovascular dis-
ease, peptic ulcers, diabetes mellitus, hypertension, or liver
disease, including inactive hepatitis B virus (HBV) car-
riers, chronic HBV or hepatitis C virus (HCV) infection,
and liver cirrhosis [13]; ICPis used, including nivolumab,
pembrolizumab, atezolizumab, durvalumab, ipilimumab, or
a combination of nivolumab and ipilimumab; concomitant
medications at ICPi initiation, including PPIs, non-steroi-
dal anti-inflammatory drugs (NSAIDs), diuretics, includ-
ing furosemide, thiazides, and spironolactone, and anti-
biotics; kidney biopsy data reviewed by a board-certified
renal pathologist (Y.O.); laboratory parameters, including
baseline sCr level, baseline estimated glomerular filtration
rate (eGFR) [14], chronic kidney disease (CKD) defined as
eGFR < 60mL/min/1.73 m2 at baseline, and dipstick urine
protein at the start of ICPi treatment (negative, ± , 1 + , 2 + ,
3 + , or 4 +); malignancy type, including lymphoma, lung,
gastric, colorectal, genitourinary, breast, and head and neck
cancers; and oral corticosteroid whose initial dose for treat-
ing ICPi-AKI was recorded. Dates of initial ICPi adminis-
tration, discontinuation, and resumption were also collected
from the medical records for each patient. All consecutive
patients receiving ICPis were included to reduce selection
bias.
Statistical analyses
Statistical analyses were performed using STATA version
15.1 software (StataCorp, College Station, TX). Continuous
variables are presented as mean and standard deviation (SD)
or median and associated interquartile range (IQRs). They
were compared using Student’s t-test or Wilcoxon rank-sum
test according to distribution. Categorical variables are pre-
sented as number and percentage, and they were compared
using a χ2 test. Univariate and multivariate logistic regres-
sion analyses were performed to identify risk factors for
ICPi-AKI and factors associated with ICPi-AKI resolution
among patients with ICPi-AKI. Odds ratios (ORs) and the
corresponding 95% CIs were calculated. The independent
variables included in the logistic regression analyses were
selected based on the findings of previous studies [48] and
our clinical experience. Age, sex, and variables in the uni-
variate analysis with a p value of < 0.05 were selected as
independent variables for the multivariate analysis.
Cox proportional hazards models were used to investi-
gate possible factors associated with mortality. Hazard ratios
(HRs) and the corresponding 95% confidence intervals (CIs)
were also calculated. Proportional hazards assumptions were
confirmed using a log–log plot of survival. Independent
variables included in the Cox proportional hazards mod-
els were selected based on the findings of previous studies
[48] and our clinical experience. ICPi-AKI incidence and
Clinical and Experimental Nephrology
1 3
resolution and variables in the univariate analysis with a
p value of < 0.1 were selected as independent variables for
the multivariate analysis. The Kaplan–Meier method was
used to estimate the incidence of death in patients with and
without ICPi-AKI and the results were compared using the
log-rank test.
Due to the limited number of patients, subgroup analyses
were not performed. Instead, sensitivity analyses were con-
ducted to assess the robustness of our results. The primary
analysis was repeated first, using only patients with lung can-
cer, second, excluding those who received pembrolizumab,
third, excluding those who had liver diseases at baseline,
and fourth, the starting point for the ICPi-AKI group was
set as the day of AKI occurrence to minimize the immortal
time bias. All the reported p values are two-sided. Signifi-
cane levels (α) were set at 0.05. Patients with missing data
were excluded from the analyses and those lost to follow-up
before death were not considered for further analysis.
Results
Study population andcharacteristics
Of the 154 potentially eligible patients who started ICPi
treatment, two were excluded as they had contrast-induced
nephropathy. None of the patients had pre-renal or post-renal
AKI. The remaining 152 patients who completed follow-
up were analyzed. The baseline characteristics of patients
according to the ICPi-AKI incidence are shown in Table1.
There were no differences in the baseline characteristics
between the ICPi-AKI and non-ICPi-AKI groups, except
for a higher proportion of liver diseases and higher num-
ber of patients treated with nivolumab and pembrolizumab
in the ICPi-AKI group. One patient (0.7%) with symptoms
compatible with tubular interstitial nephritis underwent renal
biopsy.
Incidence andrisk factors ofICPi‑AKI
Twenty-seven patients (18%) developed an ICPi-AKI event
at a median of 101 (IQR, 31–158) days after the initiation of
ICPi treatment: 17 patients (63%) with stage 1, eight (30%)
with stage 2, and two (7%) with stage 3. We performed
logistic regression analyses using clinical data, including
age, sex, liver disease, types of ICPis (nivolumab, pembroli-
zumab, atezolizumab, duralumab, and ipilimumab), dipstick
urine protein > 1 + , liver diseases, PPIs, ACCI, baseline sCr,
and CKD to evaluate risk factors for AKI (Table2). The
univariate analyses showed that liver disease was associ-
ated with a higher risk of ICPi-AKI development, but PPIs
were not. The multivariate analysis confirmed that liver dis-
ease was significantly associated with a higher risk of AKI.
Figure1 shows the proportion of patients with AKI stages
by ICPi type. Patients treated with pembrolizumab showed
a significantly higher ICPi-AKI incidence than those treated
with nivolumab (p = 0.009).
Mortality andpredictors ofdeath
Of the enrolled patients, 85 (59%) died during the follow-up.
Of the 27 patients with ICPi-AKI, 17 (63%) died during the
follow-up with death occurring at a median of 18days (IQR
0–194) after ICPi-AKI development. As shown in Fig.2, the
probability of survival in the ICPi-AKI and non-ICPi-AKI
groups was not significantly different. The univariate analy-
ses revealed that the presence of peritoneal metastasis was
associated with a higher risk of death. Sex and baseline sCr
level were associated with a lower risk of death in patients
receiving pembrolizumab than in those receiving nivolumab.
In contrast, the ICPi-AKI incidence, ICPi-AKI resolution,
CKD, and liver diseases were not associated with the risk of
death. The multivariate analysis adjusted for the ICPi-AKI
incidence revealed that ICPi-AKI resolution, sex, baseline
sCr level, CKD, the presence of peritoneal metastasis, and
ICPi type were consistently associated with a lower mortal-
ity risk in patients receiving pembrolizumab than in those
receiving nivolumab (Table2).
ICPi‑AKI resolution
Among the patients with ICPi-AKI, 19/27 (73%) showed
resolution.
Sensitivity analyses
When only patients with lung cancer were considered, the
association between ICPi-AKI and mortality (all causes) was
mostly unchanged, with the analysis only failing to detect a
significant difference (log-rank test, p = 0.486). Addition-
ally, the results did not change when we excluded patients
receiving pembrolizumab from the analysis (log-rank test,
p = 0.415) and patients with liver diseases at baseline (log-
rank test, p = 0.467). Finally, the association did not change
when the starting point was set as the day of AKI occurrence
for the ICPi-AKI group (log-rank test, p = 0.714) (Table3).
Discussion
The key findings of our study are as follows. First, we iden-
tified the presence of liver diseases as an independent risk
factor for ICPi-AKI. To our knowledge, this is the first study
on liver diseases as a predictor of ICPi-AKI incidence in
a cohort of Japanese patients receiving ICPis. The under-
lying mechanisms are likely multifactorial. One possible
Clinical and Experimental Nephrology
1 3
Table 1 Baseline characteristics of the study participants according to ICPi-AKI
ICPi-AKI immune checkpoint inhibitor-associated acute kidney injury, sCr serum creatinine, eGFR estimated glomerular filtration rate, CKD
chronic kidney disease, ACCI age-adjusted Charlson comorbidity index, HBV hepatitis B virus, HCV hepatitis C virus, NSAIDs non-steroidal
anti-inflammatory drugs, PPIs proton pump inhibitors, SD standard deviation
All (n = 152) Non-ICPi-AKI (n = 125) ICPi-AKI (n = 27) p value
Age (years ± SD) 67 ± 10 67 ± 10 67 ± 7 0.638
Women, no. (%) 38 (25) 30 (24) 8 (30) 0.713
Renal biopsy, no. (%) 1 (1) 0 (0) 1 (4) 0.641
Body mass index (kg/m2 ± SD) 22 ± 3 22 ± 3 22 ± 3 0.977
Baseline sCr [mg/dL; median (25%, 75%)] 0.78 (0.67, 0.93 0.78 (0.67, 0.94) 0.77 (0.57, 0.9) 0.321
eGFR [mL/min/1.73 m2; median (25%, 75%)] 72 (55, 87) 71 (54, 84) 76 (58, 100) 0.181
CKD, no. (%) 46 (30) 38 (30) 8 (30) 1.00
Dipstick urine protein, no. (%) 109 (72) 91 (73) 18 (67) 0.521
Negative, no. (%) 55 (51) 45 (50) 10 (55) 0.829
± , no. (%) 28 (26) 26 (29) 2 (11) 0.242
1 + , no. (%) 17 (16) 14 (15) 3 (17) 1.00
2 + , no. (%) 7 (6) 6 (7) 3 (17) 0.157
3 + , no. (%) 1 (1) 1 (1) 0 (0) 1.00
4 + , no. (%) 1 (1) 1 (1) 0 (0) 1.00
ACCI, points [median (25%, 75%)] 6 (6, 7) 6 (6, 7) 6 (6, 7) 0.728
Congestive heart failure, no. (%) 3 (2) 3 (2) 0 (0) 0.960
Cerebrovascular disease, no. (%) 13 (9) 12 (10) 1 (4) 0.539
Peptic ulcer disease, no. (%) 22 (14) 18 (14) 4 (15) 1.00
Liver disease, no. (%)
Inactive HBV carrier, no. (%)
Chronic HBV infection, no. (%)
Chronic HCV infection, no. (%)
Liver cirrhosis, no. (%)
7 (5)
2 (1)
0 (0)
3 (3)
2 (1)
2 (2)
2 (2)
0 (0)
0 (0)
0 (0)
5 (19)
0 (0)
0 (0)
3 (11)
2 (7)
< 0.001
Diabetes mellitus, no. (%) 27 (18) 19 (15) 8 (30) 0.133
Hypertension, no. (%) 79 (52) 61 (49) 18 (67) 0.141
Furosemide, no. (%) 18 (12) 16 (13) 2 (7) 0.647
Thiazides, no. (%) 3 (2) 2 (1.5) 1 (4) 1.00
Spironolactone, no. (%) 8 (5) 6 (5) 2 (7) 0.940
NSAIDs, no. (%) 50 (33) 39 (31) 11 (41) 0.465
PPIs, no. (%) 66 (43) 52 (42) 14 (52) 0.447
Antibiotics, no. (%) 5 (3) 3 (2) 2 (7) 0.467
Nivolumab, no. (%) 79 (52) 71 (57) 8 (30) 0.01
Pembrolizumab, no. (%) 55 (35) 38 (30) 17 (63) 0.001
Atezolizumab, no. (%) 10 (7) 8 (6) 2 (7) 0.848
Durvalumab, no. (%) 8 (5) 8 (7) 0 (0) 0.173
Ipilimumab, no. (%) 2 (1) 2 (1.5) 0 (0) 0.508
Nivolumab and Ipilimumab, no. (%) 2 (1) 1 (1) 1 (4) 0.875
Lung cancer, no. (%) 95 (63) 74 (59) 21 (78) 0.071
Gastric cancer, no. (%) 25 (16) 22 (18) 3 (11) 0.409
Colorectal cancer, no. (%) 1 (1) 1 (1) 0 (0) 0.641
Genitourinary cancers, no. (%) 20 (13) 18 (14) 2 (7) 0.330
Breast cancer, no. (%) 1 (1) 1 (1) 0 (0) 0.641
Lymphoma, no. (%) 1 (1) 1 (1) 0 (0) 0.641
Head and neck cancers, no. (%) 9 (6) 8 (6) 1 (4) 0.590
Oral prednisolone, no. (%) 15 (10) 15 (56)
Oral prednisolone initial dose [mg/day; median
(25%, 75%)]
30 (20, 30) 30 (20, 30)
Clinical and Experimental Nephrology
1 3
mechanism is the concomitant use of ICPis and sofosbu-
vir-based antiviral therapy against chronic HCV infection,
which may have predisposed patients to develop ICPi-AKI.
For instance, several observational studies have reported
that sofosbuvir-based treatment may cause AKI, presum-
ably due to tubulointerstitial nephritis [15, 16]. We specu-
late that blocking the PD1 and CTLA4 pathways with ICPis
may have induced a loss of tolerance of memory T cells and
activated drug-specific T cells, predisposing the patient to
develop ICPi-AKI when concomitantly treated with sofosbu-
vir-based antivirals [5, 6, 17]. Another potential mechanism
was synergistic tubulointerstitial damage caused by ICPis
and cirrhosis-induced ammoniagenesis. Available evi-
dence suggests that untreated metabolic acidosis can cause
Table 2 Clinical factors
associated with the ICPi-AKI
incidence
ICPi-AKI immune checkpoint inhibitor-associated acute kidney injury, PPIs proton pump inhibitors, ACCI
age-adjusted Charlson comorbidity index, sCr serum creatinine, CKD chronic kidney disease, CI confi-
dence interval
Univariate analysis Multivariate analysis
Odds ratio 95% CI p value Odds ratio 95% CI p value
Age ≥ 65 1.36 0.51–3.65 0.541 0.88 0.30–2.62 0.823
Sex 0.75 0.30–1.89 0.541 0.85 0.30–2.46 0.766
Liver disease 13.9 2.55–76.6 0.002 11.1 1.82–67.6 0.009
Nivolumab Reference Reference
Pembrolizumab 3.86 1.52–9.77 0.004 3.80 1.39–10.3 0.009
Atezolizumab 2.16 0.39–11.9 0.384 1.67 0.26–10.7 0.586
Durvalumab
Ipilimumab
Dipstick urine protein > 1 + 1.50 0.54–4.18 0.438
PPIs 1.51 0.66–3.48 0.332
ACCI 0.99 0.63–1.55 0.948
Baseline sCr 0.26 0.04–1.67 0.156
CKD 0.96 0.39–2.39 0.937
Fig. 1 Proportion of patients with different stages of AKI according to the type of immune checkpoint inhibitor. ICPi-AKI immune checkpoint
inhibitor-associated acute kidney injury, AKI acute kidney injury. ⁑p < 0.05
Clinical and Experimental Nephrology
1 3
ammoniagenesis in the kidney, stimulating complement acti-
vation and deposition of complement C and complement
C5b-9, which can induce tubulointerstitial damage [18]. We
speculate that liver cirrhosis may increase systemic ammonia
Fig. 2 Kaplan–Meier curves showing mortality in the ICPi-AKI and non-ICPi-AKI groups of patients. ICPi-AKI, immune checkpoint inhibitor-
associated acute kidney injury
Table 3 Clinical factors
affecting all causes of mortality
ICPi-AKI immune checkpoint inhibitor-associated acute kidney injury, sCr serum creatinine, CKD chronic
kidney disease, ACCI age-adjusted Charlson comorbidity index, CI confidence interval
Univariate analysis Multivariate analysis
Hazard ratio 95% CI p value Hazard ratio 95% CI p value
ICPi-AKI incidence 0.92 0.53–1.59 0.765 0.85 0.45–1.61 0.628
ICPi-AKI resolution 2.73 0.55–13.5 0.217 7.07 0.59–84.8 0.123
Sex 0.58 0.36–0.93 0.023 0.81 0.44–1.49 0.505
Baseline sCr 0.29 0.12–0.70 0.006 0.36 0.07–1.79 0.212
CKD 0.64 0.39–1.05 0.078 0.92 0.42–2.02 0.841
Peritoneal metastasis 2.17 1.13–4.16 0.019 1.41 0.71–2.78 0.325
Dipstick urine protein > 1 + 0.97 0.55–1.73 0.925
Liver disease 0.32 0.08–1.32 0.116
ACCI 1.01 0.81–1.26 0.931
Nivolumab Reference Reference
Pembrolizumab 0.46 0.28–0.77 0.003 0.53 0.31–0.90 0.02
Atezolizumab 0.55 0.20–1.51 0.245 0.65 0.23–1.82 0.413
Durvalumab
Ipilimumab 2.00 0.27–14.7 0.498 1.41 0.71–2.78 0.325
Clinical and Experimental Nephrology
1 3
concentrations and trigger complement activation through
a similar mechanism, resulting in renal tubulointerstitial
damage. Thus, ICPis and liver cirrhosis may synergistically
contribute to the development of ICPi-AKI. However, our
findings could not prove these speculations as we did not
evaluate ammonia concentration in our data set and just one
patient underwent a kidney biopsy. Consistent with the find-
ings of previous studies [5, 6], we advocate the use of kidney
biopsies to accurately determine the cause of AKI in patients
receiving ICPis. Furthermore, we found that pembrolizumab
use was associated with a higher risk for ICPi-AKI. This
was not unexpected as the finding was consistent with that
of previous randomized-controlled trials [1921]. Although
the exact mechanism of ICPi-induced nephrotoxicity has not
been fully defined, previous studies provide insights into this
issue, including the direct binding of ICPis to the surface
proteins of kidney cells, cross-reactivity of activated T-cells
with the kidney tissue, overproduction of pro-inflammatory
cytokines, and autoantibody formation [1]. Clearly, further
studies are needed to fully understand the underlying mecha-
nisms, which is essential to develop strategies to reduce the
risk of ICPi-AKI.
Second, ICPi-AKI did not affect mortality, which was
confirmed by the results of the sensitivity analyses. This is
not consistent with the findings of previous studies, which
reported that AKI increases mortality risk [7, 8]. A pos-
sible explanation for this discrepancy is our study may not
have had the statistical power to detect differences, as only
18% of our patients developed ICPi-AKI. Close monitoring
and intensive management of patients by experienced medi-
cal professionals may improve the survival of patients with
ICPi-AKI [22]. Regardless, given its rapid onset and high
mortality rate, our results suggest the necessity of timely rec-
ognition and management of this complication. For instance,
machine-learning prediction models and/or electronic AKI
alerts may prove beneficial. In a recent observational study, a
machine-learning prediction model accurately predicted the
risk of AKI [23]. Other studies have also shown that clinical
decision support systems and electronic AKI alerts contrib-
ute to improved outcomes of patients with AKI [2426].
We anticipate future studies to evaluate whether they can
be successfully applied to in the management of ICPi-AKI.
Third, 70% of patients with ICPi-AKI showed renal func-
tion recovery, although ICPi-AKI resolution was not asso-
ciated with patient survival even after the adjustment for
covariates. Of note, our cohort presented a higher ICPi-AKI
recovery rate than the cohorts in a recent study [6]. This
can be attributed to the fact that 63% of our patients with
ICPi-AKI had stage 1 AKI and may have been more likely
to recover from AKI than those with more severe stages of
AKI. This may also be explained by the fact that ICPis were
withdrawn by all patients with ICPi-AKI and only two of the
patients resumed ICPi treatment after complete resolution
of their ICPi-AKI. While a guideline recommends gluco-
corticoid therapy [27], the results of previous observational
studies are controversial [28, 29]. Investigating whether this
particular patient population may benefit from glucocorti-
coid therapy is a future research interest.
Finally, we could not demonstrate an association between
PPIs and ICPi-AKI, inconsistent with the findings of previ-
ous studies [5, 6]. In our cohort, PPIs were used by over 60%
of the patients at the time of ICPi initiation, comparable
with the proportion in previous studies [5, 6]. The reason
for the inconsistency remains uncertain, but it may be due
to racial differences or related to the duration and/or dosage
of PPIs used.
Our study had some limitations. First, the generalizabil-
ity of our findings may be limited as our study population
comprised only Japanese patients. Second, given its obser-
vational design, residual confounding factors may exist.
Future studies using prospective enrollment and data col-
lection from the initiation of ICPis should be performed to
validate our findings. Third, non-protocolized collection
of laboratory data, such as the lack of controlled intervals,
may cause ascertainment bias, and it is possible that we
missed several AKI cases. Fourth, our multivariable mod-
els may have been underpowered and potential associations
may have been missed owing to the low rates of primary
outcome events. Fifth, it is noteworthy when interpreting
our results that only one patient underwent kidney biopsy
to determine the cause of AKI. Sixth, there is a possibility
that some patients in the ICPi-AKI group might have had
alternative etiologies of AKI because just one patient under-
went renal biopsy. Finally, owing to the small number of
patients, our study was not adequately powered to detect all
potentially significant differences; however, we conducted
this study using the maximum available number of patients
in our medical center.
Even with these limitations, our study had several
strengths. First, we were able to follow-up all patients
throughout the study period. This contributed to the com-
pleteness of the data and overcame weaknesses of other mul-
ticenter studies, where selective laboratory testing may have
led to missing follow-up data. Second, we included patients
with stage 1 AKI because a previous study proved that even
a small increase in the sCr level is associated with higher
mortality [7].
Overall, we determined that ICPi-AKI did not affect mor-
tality in patients receiving ICPis. Furthermore, liver diseases
and pembrolizumab use were independent risk factors for
developing ICPi-AKI in Japanese patients. Even with the
small number of patients, our study is significant because
it is the first to evaluate ICPi-AKI in an Asian population.
Our study has several clinical and research-related implica-
tions that will be beneficial in improving the care of patients
with cancer treated with ICPis. Our findings warrant future
Clinical and Experimental Nephrology
1 3
multi-center investigations on optimal management strate-
gies for ICPi-AKI.
Acknowledgments We would like to thank Editage (www.edita ge.jp)
for English language editing.
Authors’ contributions Conceptualization: YS and HT; Methodology:
YS, SW, KA, and TM; Formal analysis and investigation: YS, YO,
and SW; Writing: original draft preparation: YS; Writing: review and
editing: HT; Supervision: HT.
Compliance with ethical standards
Conflict of interest The authors have declared that no conflict of inter-
est exists.
Ethical approval All procedures performed in studies involving human
participants were in accordance with the ethical standards of the insti-
tutional and/or national research committee at which the studies were
conducted (IRB approval number 2-019096-00) and with the 1964
Helsinki declaration and its later amendments or comparable ethical
standards.
Informed consent Informed consent was obtained from all individual
participants included in the study.
References
1. Sury K, Parezella MA, Shirali AC. Cardiorenal complications of
immune checkpoint inhibitors. Nat Rev Nephrol. 2018;14:571–88.
2. Cortazar FB, Marrone KA, Troxell ML, Ralto KM, Hoenig MP,
Brahmer JR, etal. Clinicopathological features of acute kidney
injury associated with immune checkpoint inhibitors. Kidney Int.
2016;90:638–47.
3. Shirali AC, Perazella MA, Gettinger S. Association of acute inter-
stitial nephritis with programmed cell death 1 inhibitor therapy in
lung cancer patients. Am J Kidney Dis. 2016;68:287–91.
4. Mamlouk O, Selament U, Machado S, Abdelrahim M, Glass WF,
Tchakarov A, etal. Nephrotoxicity of immune checkpoint inhibi-
tors beyond tubulointerstitial nephritis: Single-center experience.
J Immunother Cancer. 2019;7:2.
5. Seethapathy H, Zhao S, Chute DF, Zubiri L, Oppong Y, Strohbehn
I, etal. The incidence, causes, and risk factors of acute kidney
injury in patients receiving immune checkpoint inhibitors. Clin J
Am Soc Nephrol. 2019;14:1692–700.
6. Cortarzar FB, Kibbelaar ZA, Glezerman IG, Budayyeh A, Mam-
louk O, Motwani SS, etal. Clinical features and outcomes of
immune checkpoint inhibitor-associated AKI: A multicenter
study. J Am Soc Nephrol. 2020;31:435–46.
7. Sawhney S, Marks A, Fluck N, Levin A, Prescott G, Black C.
Intermediate and long-term outcomes of survivors of acute kid-
ney injury episodes: a large population-based cohort study. Am J
Kidney Dis. 2017;69:18–28.
8. Fujii T, Uchino S, Doi K, Sato T, Kawamura T, JAKID study
group. Diagnosis, management, and prognosis of patients with
acute kidney injury in Japanese intensive care units: the JAKID
study. J Crit Care. 2018;47:185–91.
9. Tabei A, Watanabe M, Ikeuchi H, Nakasatomi M, Sakairi T,
Kaneko Y, etal. The analysis of renal infiltrating cells in acute
tubulointerstitial nephritis induced by anti-PD-1 antibodies: a case
report and review of literature. Intern Med. 2018;57:3135–9.
10. The STROBE checklist. http://www.plosm edici ne.org/.
Accessed 6 Mar 2020
11. Kellum JA, Lameire N, Aspelin P, Barsoum RS, Burdmann
EA, Goldstein SL, etal. Kidney disease: Improving Global
Outcomes (KDIGO) acute kidney injury work group. KDIGO
clinical practice guideline from acute kidney injury. Kidney Int
Suppl. 2012;2:1–138.
12. Sndararajan V, Henderson T, Perry C, Muggivan A, Quan H,
Ghali WA. New ICD-10 version of the Charlson comorbid-
ity index predicted in-hospital mortality. J Clin Epidemiol.
2004;57:1288–94.
13. Goldman L, Schafer AI. Chronic viral and autoimmune hepati-
tis. In: Goldman-Cecil Medicine. 25th ed. Philadelphia: Elsevier
Saunders; 2014. p. 993–1006.
14. Matsuo S, Imai E, Horio M, Yasuda Y, Tomita K, Nitta K, etal.
Collaborators developing the Japanese equation for estimated
GFR Revised equations for estimated GFR from serum creatinine
in Japan. Am J Kidney Dis. 2009;53:982–92.
15. Maan R, Al Marzooqi SH, Klair JS, Karkada J, Cerocchi O, Kow-
gier M, etal. The frequency of acute kidney injury in patients with
chronic hepatitis C virus infection treated with sofosbuvir-based
regimens. Aliment Pharmacol Ther. 2017;46:46–55.
16. Brown PR, Sadiq O, Weick A, Lenhart A, Elbatta M, Fernan-
dez C, etal. Acute kidney injury in patients undergoing chronic
hepatitis C virus treatment with Ledipasvir/Sofosbuvir. Hepatol
Commun. 2018;24:1172–8.
17. Gibson A, Gaulkner L, Lichtenfels M, Ogese M, Park BK, Naisbitt
DJ. The effect of inhibitory signals on the priming of drug hapten-
specific T cells that express distinct Vβ receptors. J Immunol.
2017;199:1223–7.
18. Wesson DE, Buysse JM, Bushinsky DA. Mechanisms of meta-
bolic acidosis-induced kidney injury in chronic kidney disease. J
Am Soc Nephrol. 2020. https ://doi.org/10.1681/ASN.20190 70677
(Accessed March 6, 2020).
19. Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han
JY, etal. Pembrolizumab versus docetaxel for previously
treated, PD-L1-positive, advanced non-small-cell lung can-
cer (KEYNOTE-010): a randomized controlled trial. Lancet.
2016;387:1540–50.
20. Reck M, Rodríguez-Abreu D, Robinson AG, Hui R, Csőszi
T, Fűlőp A, etal. Pembrolizumab versus chemotherapy for
PD-L1-positive non-small-cell lung cancer. N Engl J Med.
2016;375:1823–33.
21. Bellmunt J, Vaughn WDJ, Lee FJL, Fong L, Vogelzang NJ,
Climent MA, etal. Pembrolizumab as second-line therapy for
advanced urothelial carcinoma. N Engl J Med. 2017;376:1015–26.
22. Rizo-Topete LM, Rosner MH, Ronco C. Acute kidney injury risk
assessment and nephrology rapid response team. Blood Purif.
2017;43:82–8.
23. Koyner JL, Carey KA, Edelson DP, Churpek MM. The develop-
ment of a machine learning inpatient acute kidney injury predic-
tion model. Crit Care Med. 2018;46:1070–7.
24. Mendu ML, Ciociolo GR Jr, Mclaughlin SR, Graham DA, Ghaz-
inouri R, Parmar S, etal. A decision-making algorithm for initia-
tion and discontinuation of RRT in severe AKI. Clin J Am Soc
Nephrol. 2017;12:228–36.
25. Park S, Baek SH, Ahn S, Lee KH, Hwang H, Ryu J, etal. Impact
of electronic acute kidney injury (AKI) alerts with automated
nephrologist consultation on detection and severity of AKI: a
quality improvement study. Am J Kidney Dis. 2018;71:9–19.
26. Al-Jaghbeer M, Dealmeida D, Bilderback A, Ambrosino R, Kel-
lum JA. Clinical decision support for in-hospital AKI. J Am Soc
Nephrol. 2018;29:654–60.
27. Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ,
Caterino JM, etal. Management of immune-related adverse events
in patients treated with immune checkpoint inhibitor therapy:
Clinical and Experimental Nephrology
1 3
American Society of Clinical Oncology clinical practice guide-
line. J Clin Oncol. 2018;36:1714–68.
28. González E, Gutiérrez E, Galeano C, Chevia C, de Sequera P,
Bernis C, etal. Early steroid treatment improves the recovery
of renal function in patients with drug-induced acute interstitial
nephritis. Kidney Int. 2008;73:940–6.
29. Fernandez-Juarez G, Perez JV, Caravaca-Fontán F, Quintana
L, Shabaka A, Rodriguez E, etal. Duration of treatment with
corticosteroids and recovery of kidney function in acute interstitial
nephritis. Clin J Am Soc Nephrol. 2018;13:1851–8.
Publisher’s Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
... Three of the four matched case-control studies [25 , 28 , 30 ] and one of the retrospective studies reported ICI-related AKI [41 ] and were therefore excluded. Thus, by including 116 studies with 44 158 patients, the pooled all-cause AKI occurrence rate was 7.4% ( 95% CI 5.8-9.0) with high heterogeneity [ I 2 = 98% ( 95% CI 98-98) ] ( Fig. 1 ) . ...
... By including 25 retrospective cohort studies with 21 568 patients, the pooled ICI-related AKI occurrence rate was 3.2% ( 95% CI 2.2-4.3) with high heterogeneity [ I 2 = 93% ( 95% CI 90-94) ] ( Fig. 2 ) . One outlier study [41 ] was noted. ...
Article
Full-text available
Background Immune checkpoint inhibitors (ICIs) have been associated with acute kidney injury (AKI). However, the occurrence rate of ICI-related AKI has not been systematically examined. Additionally, exposure to proton pump inhibitors (PPIs) and non-steroidal anti-inflammatory drugs (NSAIDs) were considered as risk factors for AKI, but with inconclusive results in ICI-related AKI. Our aim was to analyse the occurrence rate of all-cause AKI and ICI-related AKI and the occurrence rates of severe AKI and dialysis-requiring AKI, and to determine whether exposure to PPIs and NSAIDs poses a risk for all-cause and ICI-related AKI. Methods This study population was adult ICI recipients. A systematic review was conducted by searching MEDLINE, Embase and PubMed through October 2023. We included prospective trials and observational studies that reported any of the following outcomes: the occurrence rate of all-cause or ICI-related AKI, the relationship between PPI or NSAID exposure and AKI development or the mortality rate in the AKI or non-AKI group. Proportional meta-analysis and pairwise meta-analysis were performed. The evidence certainty was assessed using the Grading of Recommendations Assessment, Development and Evaluation framework. Results A total of 120 studies comprising 46 417 patients were included. The occurrence rates of all-cause AKI were 7.4% (14.6% from retrospective studies and 1.2% from prospective clinical trials). The occurrence rate of ICI-related AKI was 3.2%. The use of PPIs was associated with an odds ratio (OR) of 1.77 [95% confidence interval (CI) 1.43–2.18] for all-cause AKI and an OR of 2.42 (95% CI 1.96–2.97) for ICI-related AKI. The use of NSAIDs was associated with an OR of 1.77 (95% CI 1.10–2.83) for all-cause AKI and an OR of 2.57 (95% CI 1.68–3.93) for ICI-related AKI. Conclusions Our analysis revealed that approximately 1 in 13 adult ICI recipients may experience all-cause AKI, while 1 in 33 adult ICI recipients may experience ICI-related AKI. Exposure to PPIs and NSAIDs was associated with an increased OR risk for AKI in the current meta-analysis.
... Occurrence rate of all-cause AKI and ICIs-related AKI CI: 90-94%) ( Figure 2). An outlier study (Shimamura, 2021) was noted. ...
... Three of the four matched case-control studies [25, 28, 30] and one of the retrospective studies only reported ICIs-related AKI[41], and were therefore excluded. Therefore, by including 116 studies with 44158 patients, the pooled all-cause AKI occurrence rate was ...
Article
Full-text available
Background It is noted immune checkpoint inhibitors (ICIs) were associated with AKI. However, the occurrence rate of ICIs-related AKI was not been systematically examined. Additionally, exposure to PPI and NSAID were considered as a risk factor for AKI but with inconclusive results in ICIs related AKI. Our aim was to analyze the occurrence rate of all-cause AKI and ICIs-related AKI, to analyze the occurrence rates of severe AKI and dialysis-requiring AKI, to determine whether exposure to PPIs and NSAIDs posed a risk factor for all-cause and ICIs-related AKI. Methods This study population was adult ICI recipients. Systematic review by searching MEDLINE, EMBASE, PubMed through Oct 2023. Prospective trials and observational studies that reported any of the following outcomes: the occurrence rate of all-cause or ICIs-related AKI, the relationship between PPI or NSAID exposure and AKI development, or the mortality rate in the AKI or non-AKI group. Proportional meta-analysis, pairwise meta-analysis was performed. The evidence certainty was assessed using GRADE framework. Results A total of 120 studies comprising 46 417 patients were included. The occurrence rates of all-cause AKI were 7.4% (14.6% from retrospective studies and 1.2% from prospective clinical trials). The occurrence rate of ICIs-related AKI was 3.2%. The use of PPIs was associated with an odds ratio (OR) of 1.92 1.77 (95% CI: 1.43–2.18) for all-cause AKI and an OR of 2.42 (95% CI: 1.96–2.97) for ICI-related AKI. The use of NSAIDs was associated with an OR of 1.77 (95% CI: 1.10–2.83) for all-cause AKI and an OR of 2.57 (95% CI: 1.68–3.93) for ICI-related AKI. Conclusions Our analysis revealed that approximately 1 in 13 adult ICIs recipients may experience all-cause AKI, while 1 in 33 adult ICI recipients may experience ICIs-related AKI. Exposure to PPIs and NSAIDs was associated with an increased odds ratio risk for AKI in current meta-analysis.
... a previous meta-analysis based on clinical trials showed an incidence of 2.2% [9]. With the increasing application of anti-PD-1/PD-l1 antibodies, the incidence of ici-related aKi, in reality, may be higher, ranging from 3-18%, as reported in the recent literature [10][11][12][13][14][15]. some studies have shown that using proton pump inhibitors (PPis), non-steroidal anti-inflammatory drugs (NsaiDs), and antibiotics at baseline is associated with the development of aKi [11,16], but others have not suggested a role for such risk factors [12,14]. ...
... in our study, the incidence of aKi associated with anti-PD-1/ PD-l1 antibodies was 6.5%, which was similar to that reported by Ji et al. [10] and sorah et al. [22]. however, previous studies have reported a higher incidence of aKi, ranging from 14.2 to 18% [12][13][14][15]. a possible reason for this difference is that we screened for the etiology of aKi and excluded hemodynamic and obstructive causes. ...
Article
Full-text available
Anti-PD-1/PD-L1 antibodies are widely used in anti-cancer therapy. While they have improved cancer prognoses, immune-related adverse events, which can cause acute kidney injury (AKI), cannot be ignored. The purpose of this retrospective cohort study was to assess the incidence, risk factors, and prognosis of AKI associated with anti-PD-1/PD-L1 antibodies. Patients who received anti-PD-1/PD-L1 antibody treatment at our hospital between January 2018 and December 2022 were enrolled. Clinical information, combined medications, concomitant diseases, tumor types, and laboratory indicators were collected from patient records, and the incidence of AKI was determined. The risk factors for AKI were assessed using univariate and multivariate logistic regression analyses. Overall, 1418 patients were enrolled. The median follow-up time was 112 days and 92 (6.5%) developed AKI. The median time from the initial anti-PD-1/PD-L1 antibody treatment to AKI was 99.85 days. Head and neck cancer and combined use of diuretics, non-steroidal anti-inflammatory drugs (NSAIDs), lower hemoglobin level, and other types of chemotherapeutic drugs were independent risk factors for AKI. The complete recovery, partial recovery, non-recovery, and unknown AKI rates were 7.6%, 28.3%, 52.2%, and 11.9%, respectively. Kidney biopsies were performed on two patients with AKI and pathology confirmed diagnosis of acute tubulointerstitial nephritis. In this cohort, AKI was not uncommon in patients treated with anti-PD-1/PD-L1 antibodies; therefore, it is necessary to monitor renal function and identify AKI early, especially in patients with head and neck tumors. Improving anemia and minimizing the use of diuretics, NSAIDs, and chemotherapeutics may reduce AKI.
... Immune checkpoint inhibitors (ICIs) are key therapeutic agents for advanced cancer that can cause renal-related adverse events during their administration, including AKI [4][5][6][7][8]. With the increasing use of ICIs, the incidence of AKI during ICI therapy has been reported to be as high as 14-18% [9][10][11]. The development of AKI during systemic therapy, such as ICI therapy, not only increases the risk of death and the adverse effects on multiple organs but also represents a major cause of interruption of cancer treatment [1,2]. ...
Article
Full-text available
Background Acute kidney injury (AKI) is a critical complication of immune checkpoint inhibitor therapy. Since the etiology of AKI in patients undergoing cancer therapy varies, clarifying underlying causes in individual cases is critical for optimal cancer treatment. Although it is essential to individually analyze immune checkpoint inhibitor-treated patients for underlying pathologies for each AKI episode, these analyses have not been realized. Herein, we aimed to individually clarify the underlying causes of AKI in immune checkpoint inhibitor-treated patients using a new clustering approach with Shapley Additive exPlanations (SHAP). Methods We developed a gradient-boosting decision tree-based machine learning model continuously predicting AKI within 7 days, using the medical records of 616 immune checkpoint inhibitor-treated patients. The temporal changes in individual predictive reasoning in AKI prediction models represented the key features contributing to each AKI prediction and clustered AKI patients based on the features with high predictive contribution quantified in time series by SHAP. We searched for common clinical backgrounds of AKI patients in each cluster, compared with annotation by three nephrologists. Results One hundred and twelve patients (18.2%) had at least one AKI episode. They were clustered per the key feature, and their SHAP value patterns, and the nephrologists assessed the clusters’ clinical relevance. Receiver operating characteristic analysis revealed that the area under the curve was 0.880. Patients with AKI were categorized into four clusters with significant prognostic differences (p = 0.010). The leading causes of AKI for each cluster, such as hypovolemia, drug-related, and cancer cachexia, were all clinically interpretable, which conventional approaches cannot obtain. Conclusion Our results suggest that the clustering method of individual predictive reasoning in machine learning models can be applied to infer clinically critical factors for developing each episode of AKI among patients with multiple AKI risk factors, such as immune checkpoint inhibitor-treated patients.
... 3 In recent observational studies, the risk of AKI was as high as 25% within the first year after initiation of ICIs. [4][5][6][7][8][9][10][11][12] Moreover, AKI in patients receiving ICIs is associated with a more than 2-fold increased mortality. 4 Proton pump inhibitors (PPIs) are widely used in the treatment of peptic ulcers and gastroesophageal reflux disease but have been associated with an increased risk of AKI. ...
Article
Full-text available
Previous studies have suggested that the use of proton pump inhibitors (PPIs) more than doubles the risk of acute kidney injury (AKI) in cancer patients receiving immune checkpoint inhibitors (ICIs). However, this association may be confounded. Therefore, we conducted a register‐based cohort study to examine the risk of AKI in users and nonusers of PPIs among cancer patients treated with ICIs in Denmark from 2011 through 2021 while accounting for a comprehensive range of potential confounders. PPI use was determined based on redeemed prescriptions of PPIs before ICI initiation. We identified laboratory‐recorded AKI events within the first year after ICI initiation. We estimated the risks and hazard ratios (HRs) of AKI while accounting for a comprehensive range of confounders (including comorbidities and comedication) by propensity score weighting. Furthermore, we performed an additional per‐protocol analysis while accounting for informative censoring by weighting. We identified 10 200 cancer patients including 2749 (27%) users, 6214 (61%) nonusers, and 1237 (12%) former users of PPIs. PPI users had an increased risk of AKI compared to nonusers (1‐year risk, 24.7% vs 19.9%; HR, 1.42 [95% confidence interval (CI), 1.29‐1.56]); however, this association attenuated when accounting for confounders (weighted 1‐year risk, 24.2% vs 23.8%; weighted HR, 1.06 [95% CI, 0.93‐1.21]). In the per‐protocol analysis, the crude HR was 1.86 (95% CI, 1.63‐2.12), while the weighted HR was 1.24 (95% CI, 1.03‐1.49). Thus, the association between PPI use and AKI could largely be explained by confounding, suggesting that previous studies may have overestimated the association.
... We demonstrate that survival was closely linked to malignancy status and differed by AKI causes. ICPi-AKI per se was not associated with an increased risk of death, a finding that is supported by other reports (13)(14)(15)(23)(24)(25)(26). In contrast, non-ICPi-AKI was associated with reduced survival in patients with treatment response. ...
Article
Background: Immune checkpoint inhibitors (ICPi) can cause immune-related adverse events (irAEs) including acute kidney injury (AKI). We investigated the incidence of ICPi-associated AKI (ICPi-AKI) and AKI from other causes (non-ICPi-AKI) in cancer patients treated with ICPi. Methods: This was a single-centre retrospective cohort study of patients receiving ICPi therapy between December 2011 and August 2020. AKI was defined and staged by the Kidney Disease Improving Global Outcomes creatinine criteria. The primary outcome was the incidence of AKI and ICPi-AKI. Results: A total of 1037 patients were included in the final analysis. The median age was 63 years, 60% were male, and 22% had pre-existing chronic kidney disease. Overall, 189 patients (18.2%) developed AKI of whom 37 patients (3.6%) had ICPi-AKI. In patients with progressive cancer, AKI was not associated with increased mortality. In treatment responders, non-ICPi-AKI was associated with an increased risk of mortality (adjusted hazard ratio [HR] 2.03; 95% confidence interval [CI] 1.12-3.67), whereas ICPi-AKI was not linked to an increased risk of death (adjusted HR 0.60; 95% CI 0.18-1.96). Patients with ICPi-AKI were more likely to have higher AKI stages and less likely to have complete kidney recovery compared with non-ICPi-AKI (54% versus 79%, p = 0.01). Conclusion: AKI was common in cancer patients treated with ICPi. Patients with ICPi-AKI had worse kidney outcomes compared to those with AKI from other causes. However, non-ICPi-AKI was associated with a higher risk of death. These findings emphasise the importance of identifying different sub-phenotypes of AKI.
... In a Japanese report, ICI-related AKI diagnosed by a nephrologist developed in 27 of 152 (18%) patients treated with ICIs, and concomitant liver disease was an independent risk factor for the development of AKI (OR 11.1, 95% CI 1.82-67.6) [273]. ...
Article
The prevalence of CKD may be higher in patients with cancer than in those without due to the addition of cancer-specific risk factors to those already present for CKD. In this review, we describe the evaluation of kidney function in patients undergoing anticancer drug therapy. When anticancer drug therapy is administered, kidney function is evaluated to (1) set the dose of renally excretable drugs, (2) detect kidney disease associated with the cancer and its treatment, and (3) obtain baseline values for long-term monitoring. Owing to some requirements for use in clinical practice, a GFR estimation method such as the Cockcroft-Gault, MDRD, CKD-EPI, and the Japanese Society of Nephrology's GFR estimation formula has been developed that is simple, inexpensive, and provides rapid results. However, an important clinical question is whether they can be used as a method of GFR evaluation in patients with cancer. When designing a drug dosing regimen in consideration of kidney function, it is important to make a comprehensive judgment, recognizing that there are limitations regardless of which estimation formula is used or if GFR is directly measured. Although CTCAEs are commonly used as criteria for evaluating kidney disease-related adverse events that occur during anticancer drug therapy, a specialized approach using KDIGO criteria or other criteria is required when nephrologists intervene in treatment. Each drug is associated with the different disorders related to the kidney. And various risk factors for kidney disease associated with each anticancer drug therapy.
Article
Full-text available
Purpose The administration of immune checkpoint inhibitors (ICIs) may lead to renal adverse events, notably including renal dysfunction. To early predict the probability of renal dysfunction after ICIs therapy, a retrospective case–control study was conducted. Methods Clinical information on ICIs-treated patients was collected. Multivariable logistic regression was applied to identify risk factors for renal dysfunction after ICIs treatment. Moreover, a nomogram model was developed and validated internally. Results A total of 442 patients were included, among which 35 (7.9%) experienced renal dysfunction after ICIs treatment. Lower baseline estimated glomerular filtration rate (eGFR) (OR 0.941; 95% CI 0.917 to 0.966; p<0.001), concurrent exposure of platinum(OR 4.014; 95% CI 1.557 to 10.346; p=0.004), comorbidities of hypertension (OR 3.478; 95% CI 1.600 to 7.562; p=0.002) and infection (OR 5.402; 95% CI 1.544 to 18.904; p=0.008) were found to be independent associated with renal dysfunction after ICIs treatment. To develop a predictive nomogram for the occurrence of renal dysfunction after ICIs treatment, the included cases were divided into training and validation groups in a ratio of 7:3 randomly. The above four independent risk factors were included in the model. The area under the receiver operating characteristic curves of the predictiive model were 0.822 (0.723–0.922) and 0.815 (0.699–0.930) in the training and validation groups, respectively. Conclusions Lower baseline eGFR, platinum exposure, comorbidities of hypertension and infection were predictors of renal dysfunction in ICIs-treated patients with cancer. A nomogram was developed to predict the probability of renal dysfunction after ICIs treatment, which might be operable and valuable in clinical practice.
Article
Introduction: Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment. However, they pose the risk of immune-related adverse events, including ICI-mediated acute kidney injury (ICI-AKI). Recent studies have implicated proton pump inhibitors (PPIs) as potential contributors to ICI-AKI development. This meta-analysis examines the association between PPI use and ICI-AKI, exploring a potential modifiable risk factor in ICI therapy, while also reviewing the possible outcomes of ICI-AKI. Methods: We conducted a comprehensive systematic review and meta-analysis of observational studies, assessing the risk of ICI-AKI in cancer patients concurrently using PPIs and potential outcomes. Odds ratios (ORs) were pooled using random-effects models. Subgroup analyses and sensitivity analyses were performed to evaluate heterogeneity and potential biases. Results: A total of 14 studies involving 12,694 patients were included. In total, we analyzed 639 patients with all-cause AKI and 779 patients with ICI-AKI. The pooled OR for the overall incidence of AKI from all-cause was 1.57 (95% Confidence Interval (CI), 1.02 to 2.40) among patients on PPIs. Specifically, the risk of ICI-AKI associated with PPI use was significantly higher, with a pooled OR of 1.84 (95% CI 1.16 to 2.90). This indicates approximately 84% higher likelihood of developing ICI-AKI with concurrent use of PPIs. Additionally, among patients with ICI-AKI, 67% had complete or partial recovery of renal function, 32% progressed to chronic kidney disease (CKD) and about 36% died during a follow-up period of at least 3 months. Conclusion: This meta-analysis highlights the importance of cautious PPI prescription in cancer patients undergoing ICI therapy. Clinicians are advised to evaluate the risks and benefits of PPI use and consider alternative therapies when feasible.
Article
Full-text available
Background The incidence and risk factors of acute kidney injury (AKI) in patients with malignancies receiving immune checkpoint inhibitors (ICIs) are being extensively reported with their widespread application. Objective This study aimed to quantify the incidence and identify risk factors of AKI in cancer patients treated with ICIs. Methods We searched the electronic databases of PubMed/Medline, Web of Science, Cochrane and Embase before 1 February 2023 on the incidence and risk factors of AKI in patients receiving ICIs and registered the protocol in PROSPERO (CRD42023391939). A random-effect meta-analysis was performed to quantify the pooled incidence estimate of AKI, identify risk factors with pooled odds ratios (ORs) and 95% confidence intervals (95% CIs) and investigate the median latency period of ICI-AKI in patients treated with ICIs. Assessment of study quality, meta-regression, and sensitivity and publication bias analyses were conducted. Results In total, 27 studies consisting of 24048 participants were included in this systematic review and meta-analysis. The overall pooled incidence of AKI secondary to ICIs was 5.7% (95% CI: 3.7%-8.2%). Significant risk factors were older age (OR: 1.01, 95% CI: 1.00–1.03), preexisting chronic kidney disease (CKD) (OR: 2.90, 95% CI: 1.65–5.11), ipilimumab (OR: 2.66, 95% CI: 1.42–4.98), combination of ICIs (OR: 2.45, 95% CI: 1.40–4.31), extrarenal immune-related adverse events (irAEs) (OR: 2.34, 95% CI: 1.53-3.59), and proton pump inhibitor (PPI) (OR: 2.23, 95% CI: 1.88–2.64), nonsteroidal anti-inflammatory drug (NSAID) (OR: 2.61, 95% CI: 1.90–3.57), fluindione (OR: 6.48, 95% CI: 2.72–15.46), diuretic (OR: 1.78, 95% CI: 1.32–2.40) and angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin-receptor blockers (ARBs) (pooled OR: 1.76, 95% CI: 1.15–2.68) use. Median time from ICIs initiation to AKI was 108.07 days. Sensitivity and publication bias analyses indicated robust results for this study. Conclusion The occurrence of AKI following ICIs was not uncommon, with an incidence of 5.7% and a median time interval of 108.07 days after ICIs initiation. Older age, preexisting chronic kidney disease (CKD), ipilimumab, combined use of ICIs, extrarenal irAEs, and PPI, NSAID, fluindione, diuretics and ACEI/ARB use are risk factors for AKI in patients receiving ICIs. Systematic review registration https://www.crd.york.ac.uk/prospero/, identifier CRD42023391939.
Article
Full-text available
Retrospective analyses and single-center prospective studies identify chronic metabolic acidosis as an independent and modifiable risk factor for progression of CKD. In patients with CKD, untreated chronic metabolic acidosis often leads to an accelerated reduction in GFR. Mechanisms responsible for this reduction include adaptive responses that increase acid excretion but lead to a decline in kidney function. Metabolic acidosis in CKD stimulates production of intrakidney paracrine hormones including angiotensin II, aldosterone, and endothelin-1 (ET-1) that mediate the immediate benefit of increased kidney acid excretion, but their chronic upregulation promotes inflammation and fibrosis. Chronic metabolic acidosis also stimulates ammoniagenesis that increases acid excretion but also leads to ammonia-induced complement activation and deposition of C3 and C5b-9 that can cause tubule-interstitial damage, further worsening disease progression. These effects, along with acid accumulation in kidney tissue, combine to accelerate progression of kidney disease. Treatment of chronic metabolic acidosis attenuates these adaptive responses; reduces levels of angiotensin II, aldosterone, and ET-1; reduces ammoniagenesis; and diminishes inflammation and fibrosis that may lead to slowing of CKD progression.
Article
Full-text available
Rationale & Objective The approved therapeutic indication for immune checkpoint inhibitors (CPIs) are rapidly expanding including treatment in the adjuvant setting, the immune related toxicities associated with CPI can limit the efficacy of these agents. The literature on the nephrotoxicity of CPI is limited. Here, we present cases of biopsy proven acute tubulointerstitial nephritis (ATIN) and glomerulonephritis (GN) induced by CPIs and discuss potential mechanisms of these adverse effects. Study design, setting, & participants We retrospectively reviewed all cancer patients from 2008 to 2018 who were treated with a CPI and subsequently underwent a kidney biopsy at The University of Texas MD Anderson Cancer Center. Results We identified 16 cases diagnosed with advanced solid or hematologic malignancy; 12 patients were male, and the median age was 64 (range 38 to 77 years). The median time to developing acute kidney injury (AKI) from starting CPIs was 14 weeks (range 6–56 weeks). The average time from AKI diagnosis to obtaining renal biopsy was 16 days (range from 1 to 46 days). Fifteen cases occurred post anti-PD-1based therapy. ATIN was the most common pathologic finding on biopsy (14 of 16) and presented in almost all cases as either the major microscopic finding or as a mild form of interstitial inflammation in association with other glomerular pathologies (pauci-immune glomerulonephritis, membranous glomerulonephritis, C3 glomerulonephritis, immunoglobulin A (IgA) nephropathy, or amyloid A (AA) amyloidosis). CPIs were discontinued in 15 out of 16 cases. Steroids and further immunosuppression were used in most cases as indicated for treatment of ATIN and glomerulonephritis (14 of 16), with the majority achieving complete to partial renal recovery. Conclusions Our data demonstrate that CPI related AKI occurs relatively late after CPI therapy. Our biopsy data demonstrate that ATIN is the most common pathological finding; however it can frequently co-occur with other glomerular pathologies, which may require immune suppressive therapy beyond corticosteroids. In the lack of predictive blood or urine biomarker, we recommend obtaining kidney biopsy for CPI related AKI.
Article
Full-text available
Ledipasvir‐sofosbuvir, a once‐a‐day, oral combination pill, was approved in 2014 for the treatment of chronic hepatitis C infection. Initial trials did not comment on nephrotoxicity; however, recent data suggest a risk of acute kidney injury (AKI) with the use of the medication. We assessed the rates of AKI in patients undergoing ledipasvir‐sofosbuvir in a large, urban tertiary care center. This single‐center retrospective observation study included all patients undergoing therapy from October 1, 2014, to October 1, 2015. Rates of AKI, defined by more than a 0.3 mg/dL increase in serum creatinine level, were calculated. Patients were followed 12 weeks after therapy to assess for sustained viral response as well as to assess for improvement of AKI after completion of therapy, defined by less than 0.2 mg/dL above baseline serum creatinine. In total, 197 patients were included in the final analysis who had completed ledipasvir‐sofosbuvir therapy and completed laboratory values. Among the patients treated, 38 (19%) had AKI during therapy. An additional 4 (2%) had AKI at the end of therapy. Of the 38 patients who experienced AKI, 20 (53%) had improvement in serum creatinine to less than 0.2 mg/dL above their baseline. When comparing for chronic kidney disease (CKD) stage, those with CKD I or II experienced AKI 17% of the time compared with 47% of the time in CKD III or worse ( P = 0.005). Conclusion: AKI was seen in nearly one‐fifth of our patients, and patients with CKD stage III or worse are at increased risk. Although ledipasvir‐sofosbuvir is generally safe in the general population, close monitoring of renal function is recommended.
Article
Full-text available
Nivolumab is an anti-programmed cell death-1 (PD-1) antibody that is utilized as an immune checkpoint inhibitor (ICI) for cancer therapy. We herein present the case of a 57-year-old man who developed acute kidney injury during treatment with nivolumab for lung cancer. A renal biopsy revealed acute tubulointerstitial nephritis. Immunohistochemical staining demonstrated marked infiltration of macrophages and T cells together with mild B cell infiltration. Of note, strong CD163+ M2 macrophage infiltration was observed. The cessation of nivolumab and high-dose prednisolone therapy improved the renal function of the patient. Further, we review the pertinent literature on renal-infiltrating cells in ICI-induced tubulointerstitial nephritis.
Article
Background: Despite increasing recognition of the importance of immune checkpoint inhibitor-associated AKI, data on this complication of immunotherapy are sparse. Methods: We conducted a multicenter study of 138 patients with immune checkpoint inhibitor-associated AKI, defined as a ≥2-fold increase in serum creatinine or new dialysis requirement directly attributed to an immune checkpoint inhibitor. We also collected data on 276 control patients who received these drugs but did not develop AKI. Results: Lower baseline eGFR, proton pump inhibitor use, and combination immune checkpoint inhibitor therapy were each independently associated with an increased risk of immune checkpoint inhibitor-associated AKI. Median (interquartile range) time from immune checkpoint inhibitor initiation to AKI was 14 (6-37) weeks. Most patients had subnephrotic proteinuria, and approximately half had pyuria. Extrarenal immune-related adverse events occurred in 43% of patients; 69% were concurrently receiving a potential tubulointerstitial nephritis-causing medication. Tubulointerstitial nephritis was the dominant lesion in 93% of the 60 patients biopsied. Most patients (86%) were treated with steroids. Complete, partial, or no kidney recovery occurred in 40%, 45%, and 15% of patients, respectively. Concomitant extrarenal immune-related adverse events were associated with worse renal prognosis, whereas concomitant tubulointerstitial nephritis-causing medications and treatment with steroids were each associated with improved renal prognosis. Failure to achieve kidney recovery after immune checkpoint inhibitor-associated AKI was independently associated with higher mortality. Immune checkpoint inhibitor rechallenge occurred in 22% of patients, of whom 23% developed recurrent associated AKI. Conclusions: This multicenter study identifies insights into the risk factors, clinical features, histopathologic findings, and renal and overall outcomes in patients with immune checkpoint inhibitor-associated AKI.
Article
Background and objectives: Immune checkpoint inhibitor use in oncology is increasing rapidly. We sought to determine the frequency, severity, cause, and predictors of AKI in a real-world population receiving checkpoint inhibitors. Design, setting, participants, & measurements: We included all patients who received checkpoint inhibitor therapy from May 2011 to December 2016 at Massachusetts General Hospital. Baseline serum creatinine, averaged 6 months before checkpoint inhibitor start date, was compared with all subsequent creatinine values within 12 months of starting therapy. AKI was defined by Kidney Disease: Improving Global Outcomes criteria for fold changes in creatinine from baseline. Sustained AKI events lasted at least 3 days and was our primary outcome. The cause of sustained AKI was determined by chart review. Cumulative incidence and subdistribution hazard models were used to assess the relationship between baseline demographics, comorbidities, and medications, and sustained AKI and potential checkpoint inhibitor-related AKI. Results: We included 1016 patients in the analysis. Average age was 63 (SD 13) years, 61% were men, and 91% were white. Mean baseline creatinine was 0.9 mg/dl (SD 0.4 mg/dl), and 169 (17%) had CKD (eGFR<60 ml/min per 1.73 m2) at baseline. A total of 169 patients (17%) experienced AKI, defined by an increase in creatinine at least 1.5 times the baseline within 12 months; 82 patients (8%) experienced sustained AKI and 30 patients (3%) had potential checkpoint inhibitor-related AKI. The first episode of sustained AKI occurred, on average, 106 days (SD 85) after checkpoint inhibitor initiation. Sixteen (2%) patients experienced stage 3 sustained AKI and four patients required dialysis. Proton pump inhibitor use at baseline was associated with sustained AKI. Conclusions: AKI is common in patients receiving checkpoint inhibitor therapy. The causes of sustained AKI in this population are heterogenous and merit thorough evaluation. The role of PPI and other nephritis-inducing drugs in the development of sustained AKI needs to be better defined.
Article
Background and objectives: Drug-induced acute interstitial nephritis represents an emerging cause of acute kidney disease, especially among polymedicated elderly patients. Although corticosteroids are frequently used, controversy exists about the timing of initiation, efficacy, safety, and duration of treatment. Design, setting, participants, & measurements: We performed a retrospective study of 182 patients with biopsy-proven drug-induced acute interstitial nephritis from 13 Spanish centers. Exposure was defined as the length of corticosteroid treatment. The main outcome was the level of serum creatinine at month 6, with respect to baseline values. Results: The most common offending agents were nonsteroidal anti-inflammatory drugs (27%). In 30% of patients, the offending drug could not be identified. The median time to suspected drug withdrawal was 11 days (interquartile range, 5-22). All patients presented with acute kidney disease and were treated with corticosteroids. The mean initial dose of prednisone was 0.8±0.2 mg/kg per day. High-dose corticosteroid treatment was maintained for 2 weeks (interquartile range, 1-4). After 6 months, the mean recovered GFR was 34±26 ml/min per 1.73 m2 and ten patients required maintenance dialysis. Use of high-dose corticosteroids for 3 weeks or treatment duration >8 weeks were not associated with better recovery of kidney function. In the multivariable analysis, delayed onset of steroid treatment (odds ratio, 1.02; 95% confidence interval, 1.0 to 1.04) and the presence of interstitial fibrosis of >50% on the kidney biopsy specimen (odds ratio, 8.7; 95% confidence interval, 2.7 to 27.4) were both associated with serum creatinine level at month 6 of >75%, with respect to baseline values. Conclusions: High-dose corticosteroid treatment for 3 weeks or prolonged treatment for >8 weeks were not associated with greater kidney function recovery in drug-induced acute interstitial nephritis. A delay in the initiation of corticosteroids resulted in worse recovery of kidney function.
Article
The development of immune checkpoint inhibitors (ICIs) has driven a revolutionary change in cancer treatment. Although traditional chemotherapeutic agents remain the first-line option for most cancers, targeted immune therapies are emerging as standard treatments for advanced-stage cancers. These agents target cell surface checkpoint proteins to stimulate the recognition and destruction of cancer cells by the immune system. Clinical studies have demonstrated these immunotherapeutics to elicit favourable antitumour responses in a variety of chemotherapy-refractory malignancies. However, use of these agents can also induce immune-related adverse events (irAEs) in off-target organs, including the heart and kidney. The most common manifestations of heart and kidney damage are myocarditis and acute interstitial nephritis, respectively, but other manifestations have been reported and, although rare, these off-target effects can be life threatening. Available data suggest that ICIs induce their off-target effects through several mechanisms, including direct binding to cell surface proteins expressed in healthy tissue, activation of T cells that cross-react with off-target tissues, generation of autoantibodies or by increasing levels of pro-inflammatory cytokines. Greater understanding of the adverse effects of cancer immunotherapies and the underlying mechanisms will facilitate the development of biomarkers to identify at-risk patients and approaches to prevent these irAEs.
Article
Purpose: To determine the proportion of diagnosis and outcomes of critically ill patients with acute kidney injury (AKI), and its association with mortality using the complete Kidney Disease Improving Global Outcomes (KDIGO) classification and Sepsis-3 definition. Methods: We conducted a multicenter prospective cohort study of 13 intensive care units (ICU) in Japan. Patients admitted to the ICUs during six months in 2016 were consecutively enrolled. Results: Among 2292 patients, AKI was diagnosed in 1024 (44.7%) patients, using the KDIGO classification. Sepsis was diagnosed in 424 patients (18.5%), of whom 281 patients (66.3%) had AKI. Septic shock was diagnosed in 166 patients (7.2%), of whom 125 patients (75.3%) had AKI. Of 1024 patients with AKI, renal replacement therapy was applied to 171 patients (16.7% of AKI) during the ICU stay. The adjusted odds ratio (aOR) of AKI to hospital mortality was 1.66 (95% confidence intervals 1.26-2.18), while that among sepsis was 0.87 (95% confidence intervals 0.55-1.37). Conclusions: AKI accounted for >40% of ICU patients with the KDIGO classification and was associated with increased risk of hospital mortality. Septic AKI was diagnosed in three-fourths of patients with sepsis, while the impact of AKI on hospital mortality among sepsis was not observed.
Article
Objectives: To develop an acute kidney injury risk prediction model using electronic health record data for longitudinal use in hospitalized patients. Design: Observational cohort study. Setting: Tertiary, urban, academic medical center from November 2008 to January 2016. Patients: All adult inpatients without pre-existing renal failure at admission, defined as first serum creatinine greater than or equal to 3.0 mg/dL, International Classification of Diseases, 9th Edition, code for chronic kidney disease stage 4 or higher or having received renal replacement therapy within 48 hours of first serum creatinine measurement. Interventions: None. Measurements and main results: Demographics, vital signs, diagnostics, and interventions were used in a Gradient Boosting Machine algorithm to predict serum creatinine-based Kidney Disease Improving Global Outcomes stage 2 acute kidney injury, with 60% of the data used for derivation and 40% for validation. Area under the receiver operator characteristic curve (AUC) was calculated in the validation cohort, and subgroup analyses were conducted across admission serum creatinine, acute kidney injury severity, and hospital location. Among the 121,158 included patients, 17,482 (14.4%) developed any Kidney Disease Improving Global Outcomes acute kidney injury, with 4,251 (3.5%) developing stage 2. The AUC (95% CI) was 0.90 (0.90-0.90) for predicting stage 2 acute kidney injury within 24 hours and 0.87 (0.87-0.87) within 48 hours. The AUC was 0.96 (0.96-0.96) for receipt of renal replacement therapy (n = 821) in the next 48 hours. Accuracy was similar across hospital settings (ICU, wards, and emergency department) and admitting serum creatinine groupings. At a probability threshold of greater than or equal to 0.022, the algorithm had a sensitivity of 84% and a specificity of 85% for stage 2 acute kidney injury and predicted the development of stage 2 a median of 41 hours (interquartile range, 12-141 hr) prior to the development of stage 2 acute kidney injury. Conclusions: Readily available electronic health record data can be used to predict impending acute kidney injury prior to changes in serum creatinine with excellent accuracy across different patient locations and admission serum creatinine. Real-time use of this model would allow early interventions for those at high risk of acute kidney injury.