Sei sulla pagina 1di 10

Research

JAMA | Original Investigation

Effect of Coaching to Increase Water Intake on Kidney


Function Decline in Adults With Chronic Kidney Disease
The CKD WIT Randomized Clinical Trial
William F. Clark, MD; Jessica M. Sontrop, PhD; Shih-Han Huang, MD, PhD; Kerri Gallo, RN; Louise Moist, MD, MSc;
Andrew A. House, MD; Meaghan S. Cuerden, MSc; Matthew A. Weir, MD, MSc; Amit Bagga, MD;
Scott Brimble, MD; Andrew Burke, MD; Norman Muirhead, MD; Sanjay Pandeya, MD; Amit X. Garg, MD, PhD

Supplemental content
IMPORTANCE In observational studies, increased water intake is associated with better CME Quiz at
kidney function. jamanetwork.com/learning

OBJECTIVE To determine the effect of coaching to increase water intake on kidney function
in adults with chronic kidney disease.

DESIGN, SETTING, AND PARTICIPANTS The CKD WIT (Chronic Kidney Disease Water Intake
Trial) randomized clinical trial was conducted in 9 centers in Ontario, Canada, from 2013 until
2017 (last day of follow-up, May 25, 2017). Patients had stage 3 chronic kidney disease
(estimated glomerular filtration rate [eGFR] 30-60 mL/min/1.73 m2 and microalbuminuria or
macroalbuminuria) and a 24-hour urine volume of less than 3.0 L.

INTERVENTIONS Patients in the hydration group (n = 316) were coached to drink more water,
and those in the control group (n = 315) were coached to maintain usual intake.

MAIN OUTCOMES AND MEASURES The primary outcome was change in kidney function (eGFR
from baseline to 12 months). Secondary outcomes included 1-year change in plasma copeptin
concentration, creatinine clearance, 24-hour urine albumin, and patient-reported overall
quality of health (0 [worst possible] to 10 [best possible]).

RESULTS Of 631 randomized patients (mean age, 65.0 years; men, 63.4%; mean eGFR,
43 mL/min/1.73 m2; median urine albumin, 123 mg/d), 12 died (hydration group [n = 5];
control group [n = 7]). Among 590 survivors with 1-year follow-up measurements (95% of
619), the mean change in 24-hour urine volume was 0.6 L per day higher in the hydration
group (95% CI, 0.5 to 0.7; P < .001). The mean change in eGFR was −2.2 mL/min/1.73 m2 in
the hydration group and −1.9 mL/min/1.73 m2 in the control group (adjusted between-group
Author Affiliations: London Health
difference, −0.3 mL/min/1.73 m2 [95% CI, −1.8 to 1.2; P = .74]). The mean between-group Sciences Centre, London, Ontario,
differences (hydration vs control) in secondary outcomes were as follows: plasma copeptin, Canada (Clark, Sontrop, Huang, Gallo,
−2.2 pmol/L (95% CI, −3.9 to −0.5; P = .01); creatinine clearance, 3.6 mL/min/1.73 m2 (95% CI, Moist, House, Cuerden, Weir,
Muirhead, Garg); Department of
0.8 to 6.4; P = .01); urine albumin, 7 mg per day (95% CI, −4 to 51; P = .11); and quality of Medicine, Western University,
health, 0.2 points (95% CI, −0.3 to 0.3; P = .22). London, Ontario, Canada (Clark,
Huang, Moist, House, Weir, Bagga,
Muirhead, Garg); Department of
CONCLUSIONS AND RELEVANCE Among adults with chronic kidney disease, coaching to
Epidemiology and Biostatistics,
increase water intake compared with coaching to maintain the same water intake did not Western University, London, Ontario,
significantly slow the decline in kidney function after 1 year. However, the study may have Canada (Sontrop, Moist, Garg);
been underpowered to detect a clinically important difference. Hôtel-Dieu Grace Hospital, Windsor,
Ontario, Canada (Bagga);
Department of Medicine, McMaster
TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT01766687. University, Hamilton, Ontario, Canada
(Brimble); Guelph General Hospital,
Guelph, Ontario, Canada (Burke);
Halton Healthcare Services, Oakville,
Ontario, Canada (Pandeya).
Corresponding Author: William F.
Clark, MD, Victoria Hospital, 800
Commissioner’s Road E, A2-343,
London, ON, Canada N6A 5W9
JAMA. 2018;319(18):1870-1879. doi:10.1001/jama.2018.4930 (william.clark@lhsc.on.ca).

1870 (Reprinted) jama.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: on 05/08/2018


Effect of Increased Water Intake on Kidney Function Decline in Chronic Kidney Disease Original Investigation Research

D
espite widespread interest, little scientific data exist
on the optimal amount of water to drink. The popu- Key Points
lar advice to “drink at least 8 glasses of water a day”
Question Does drinking more water protect against declining
originated not from primary research, but from the US Food kidney function in patients with chronic kidney disease?
and Nutrition Board in 1945, which recommended a daily
Findings In this randomized clinical trial that included 631 adult
water intake of 2.5 L per day and stated that the majority of
patients with chronic kidney disease, the 1-year decline in
this intake could come from food sources.1,2 While many
estimated glomerular filtration rate did not significantly differ
claims about the benefits of increased water intake remain between patients who were coached to drink more water
untested,2-4 a growing body of evidence suggests that drink- compared with patients who were coached to maintain their usual
ing more water may benefit the kidneys.5,6 In animals that intake (−2.2 vs −1.9 mL/min per 1.73 m2).
undergo a near complete bilateral nephrectomy, drinking
Meaning Coaching to increase water intake did not significantly
more water suppresses the plasma concentration of vaso- slow the decline in kidney function in patients with chronic kidney
pressin and improves creatinine clearance,7,8 and in studies disease at 1-year follow-up.
of humans, drinking more water is associated with better kid-
ney function and a reduced risk of kidney stones. 9 -1 3
Whether increased water intake can benefit patients with
chronic kidney disease remains unknown. Patients, Setting, and Location
In a 6-week pilot randomized clinical trial of 29 patients Adult patients with stage 3 chronic kidney disease were
with stage 3 chronic kidney disease, coaching patients to recruited from 9 Canadian chronic kidney disease clinics
drink at least 1 additional liter of water per day (in addition to across southwestern Ontario: London (3 centers), Oakville
usual fluid intake) resulted in reduced vasopressin secretion (2 centers), Windsor (2 centers), Guelph (1 center), and
(as assessed by the plasma concentration of copeptin, a gly- Hamilton (1 center). Potentially eligible patients were invited
cosylated peptide that is co-released with vasopressin from by their nephrologist to speak with a research assistant who
the hypothalamus) compared with patients in the control explained the study, confirmed eligibility (eTable 3 in
group who were coached to maintain usual fluid intake.14,15 Supplement 3), and obtained written informed consent.
Guided by this pilot study, the present 9-center Chronic Inclusion criteria were stage 3 chronic kidney disease (eGFR
Kidney Disease Water Intake Trial (CKD WIT) was conducted 30 to 60 mL/min per 1.73 m2) and an albumin-to-creatinine
to determine if coaching patients to drink more water slowed ratio of greater than 25 mg/g (2.8 mg/mmol) for women or
their decline in kidney function over 1 year compared with greater than 18 mg/g (2.0 mg/mmol) for men (from a random
those in the control group who were coached to maintain spot urine sample) or trace protein or greater (from a urine
usual fluid intake. dipstick).18 Exclusion criteria included self-reported fluid
intake of 10 cups per day or more, a 24-hour urine volume of
3 L or more, a history of kidney stones in the past 5 years,
currently taking lithium or a diuretic above a certain dose
Methods (eTable 3 in Supplement 3), or a prescribed fluid restriction of
Study Design and Oversight less than 1.5 L per day.
A parallel-group randomized clinical trial was conducted from After providing written informed consent, patients pro-
April 2013 to June 2017. Ethical approval to conduct this trial vided a prerandomization 24-hour urine sample to confirm
was obtained from Western University’s health sciences re- their urine volume was less than 3 L per day (which was an eli-
search ethics board. All patients provided written informed gibility requirement for randomization).
consent. The original protocol (Supplement 1) and the statis-
tical analysis plan (Supplement 2) for this trial are available on- Randomized Allocation
line, the updated protocol was published previously,16 and Once study eligibility was confirmed, a research assistant
changes to the original protocol are summarized in eTable 1 in contacted each patient by telephone to complete the ran-
Supplement 3. Prespecified and post hoc outcomes are sum- domization, and concealed randomized allocation was
marized in eTable 2 in Supplement 3 along with the date that implemented by computer-generated randomization while
each outcome was added. Study reporting follows recom- the patient was on the phone. The randomized sequence
mended guidelines for randomized trials of nonpharmaco- was created by an independent statistician using SAS statis-
logic treatments.17 tical software version 9.3 and was stratified by center and
Trial conduct and safety were monitored by an indepen- sex with a 1:1 allocation using random permuted block sizes
dent data and safety monitoring board, which received a of 4 and 6.
descriptive summary of trial data and adverse events at After randomization, research staff and patients were
intervals of 3 to 9 months, with no planned interim statisti- made aware of group assignment; however, the primary out-
cal analysis of the primary or secondary outcomes. The data come was an objective measure, outcome assessors (techni-
and safety monitoring board requested and received access cians performing the laboratory measurements for the pri-
to data on estimated glomerular filtration rate (eGFR) and mary and secondary outcomes) were unaware of the random
group assignment but did not discuss these data with the allocation, and the trial statistician was blinded to patient
trial investigators. allocation for the primary analysis.

jama.com (Reprinted) JAMA May 8, 2018 Volume 319, Number 18 1871

© 2018 American Medical Association. All rights reserved.

Downloaded From: on 05/08/2018


Research Original Investigation Effect of Increased Water Intake on Kidney Function Decline in Chronic Kidney Disease

Intervention Group [best possible]; item 22 from the RAND Kidney Disease
Patients in the hydration group were coached to increase Quality of Life Short Form).24,25
their water intake by 1.0 to 1.5 L per day (depending on sex
and weight), over and above usual consumed beverages, for Outcomes
the duration of the 1-year trial (eTable 4 in Supplement 3). The prespecified primary outcome was 1-year change in eGFR
The target water intake was informed by prior literature,9,19-21 from enrollment (prerandomization) to 12 months after
patient safety, and what proved feasible in the pilot trial.14,15 randomization.26,27 Prespecified secondary outcomes were
A gradual increase in water intake over 2 weeks was advised. 1-year change in plasma copeptin concentration,28,29 creati-
During week 1, patients were instructed to drink an addi- nine clearance, 24-hour urine albumin, 30 and patient-
tional cup of water at breakfast, lunch, and dinner and to reported overall quality of health (Supplement 1 and the pub-
increase to the full amount in week 2. Patients in the hydra- lished protocol16).24,25
tion group also received reusable drinking containers and Other exploratory outcomes (some prespecified and some
were mailed 20 vouchers per month, each redeemable for post hoc; eTable 2 in Supplement 3) were 1-year change in
1.5 L of bottled water. 24-hour urine osmolality, creatinine, urea, sodium, and po-
tassium; serum osmolality, creatinine, and urea; blood pres-
Control Group sure, body weight, waist circumference, and body mass in-
Patients in the control group were coached to continue with dex (calculated as weight in kilograms divided by height in
their usual fluid intake or to decrease intake by 0.25 to 0.5 L meters squared); and the 6-month change in dietary intake of
per day (1 to 2 cups/d) if their prerandomization 24-hour urine protein and sodium.
volume was greater than 1.5 L per day and 24-hour urine
osmolality was less than 500 mOsm/kg.14 Sample Size
In a previous longitudinal study of healthy adults free of
Intervention Adherence and Monitoring chronic kidney disease, those with urine volumes of 3 L per
To encourage adherence to the assigned intervention and to day or more had a significantly slower eGFR decline than
minimize cross-group contamination, patients in the hydra- those with smaller urine volumes (difference, 0.6 mL/min
tion and control groups were individually coached over the per 1.73 m2 per year; P = .01).9 Given that in previous studies
phone at monthly intervals for 12 months after randomiza- of patients with chronic kidney disease the mean decline in
tion using a standardized survey with reference to quantity eGFR ranged from 1.5 to 3.5 mL/min per 1.73 m2 (SD range,
of fluid ingested relative to the target intake. Coaching also 3-5) per year,31-33 a minimal clinically important difference
included a discussion of urine color charts (showing the of 1 mL/min per 1.73 m2 was prespecified (a small to moder-
spectrum of diluted to concentrated urine). Intervention ate effect size) and the target enrollment was 700 patients
adherence was assessed by 24-hour urine volume (collected (350 per group; assuming a standard deviation of <5; 2-sided
at 6 and 12 months after randomization and measured α = .05; power, 80%).
by a local laboratory). Adherence was also assessed by After 3.1 years of recruitment (1.3 years longer than
self-reported fluid intake (measured at 3, 6, and 9 months originally planned), 631 patients were randomized. Enroll-
after randomization). ment was stopped at this time in order to stay within the study’s
allocated budget (the trial took longer to conduct and cost
Data Collection and Measurement was more than originally estimated due to a higher than ex-
All patients, irrespective of their randomized group assign- pected screen failure rate and the need to expand into 7 addi-
ment, had the same data collection and measurement tional centers [eTable 1 in Supplement 3]). The investigators
schedule (eTable 5 in Supplement 3); more information is made this decision in consultation with the data and safety
provided in Supplement 1 and in the published protocol.16 monitoring board to ensure that patients’ contribution to this
Serum creatinine was measured using the isotope dilution trial would not be compromised by stopping the trial just short
mass spectroscopy–traceable enzymatic method, and the of the enrollment target. One year after the last patient was ran-
eGFR was calculated using the Chronic Kidney Disease Epi- domized, 12-month eGFR values for 590 patients (295 per
demiology Collaboration (CKD-EPI) equation. 22 Plasma group) were available, which enabled detection of a between-
copeptin concentration was measured from 150-μL blood group difference of at least 2 mL/min per 1.73 m2 in the mean
samples at enrollment (prerandomization), at the pa- change in eGFR over 1 year (based on an observed standard de-
tients’ next kidney care clinic visit (usually 6 to 7 months viation of 9 [2-sided α = .05; power, 80%]). No interim analy-
after enrollment), and at 12 months after randomization; ses of primary or secondary outcomes were performed.
these samples were stored at −80°C and analyzed in batches
using the automated immunofluorescent Copeptin proAVP Analysis
assay on the KRYPTOR compact PLUS (BRAHMS GmbH).23 Continuous variables are summarized as mean (SD) or as
Albuminuria was measured from a 24-hour urine sample median (interquartile range [IQR]) as appropriate. No statis-
using a standard collection jug within 2 weeks of enrollment tical tests were used to compare baseline characteristics.34
(prerandomization) and again at 6 and 12 months after ran- For the analysis of the primary outcome, the between-group
domization. Patient-reported quality of health was mea- difference in eGFR change (hydration minus control) was
sured on a 10-point Likert scale (0 [worst possible] to 10 estimated using linear regression with adjustment for the

1872 JAMA May 8, 2018 Volume 319, Number 18 (Reprinted) jama.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: on 05/08/2018


Effect of Increased Water Intake on Kidney Function Decline in Chronic Kidney Disease Original Investigation Research

following prespecified covariates measured at baseline: age in the Figure. Of 2390 patients assessed for eligibility, 1568
(in years), sex, obesity (body mass index ≥30), current did not meet the study’s eligibility criteria or did not wish to
smoker (yes/no), presence of diabetes, and 24-hour urine participate. Of 822 patients who consented, 191 were
albumin (mg/d; log-transformed); and use of an angiotensin- excluded before randomization: 76 withdrew for personal
converting enzyme inhibitor or angiotensin receptor blocker, reasons, 2 due to physician decision, 1 died, 47 had a 24-hour
diuretic, β-blocker, calcium channel blocker, or statin. 35 urine volume above 3 L, 31 failed to provide a 24-hour urine
Missing baseline data occurred for less than 0.2% of cat- sample, and 34 were ineligible for other reasons (eg, medica-
egorical covariates (if missing, the condition was considered tion contraindications or chemotherapy). The remaining 631
absent) and was 6% for 24-hour urine albumin (imputed patients were randomized to the hydration group (n = 316) or
using fully conditionally specified models36 [eAppendix 1 in the control group (n = 315). During the 1-year trial period, 12
Supplement 3]). Patients who died within 1 year of follow-up of 631 patients died (1.9%) (5 [1.6%] in the hydration group
were excluded from the primary and secondary outcome and 7 [2.2%] in the control group); of the 619 survivors, 95%
analyses (12 of 631 patients [1.9%]). Less than 5% of survi- provided 1-year follow-up measurements (last day of follow-
vors were missing a 12-month eGFR value (±4 months); in up, May 25, 2017).
the primary analysis only, missing eGFR data were imputed Baseline characteristics of 631 randomized patients are
using fully conditionally specified models36 (eAppendix 1 in shown in Table 1. There were no clinically important differ-
Supplement 3). ences between randomized groups. Patients had a mean
Several supplementary analyses of eGFR change were con- (SD) age of 65.0 years (11.8); 63.4% were men, 90.6% were
ducted using nonimputed data: (1) eGFR measured with cys- white, 45.6% were obese (body mass index ≥30), 88.1% had
tatin C; (2) a longitudinal analysis of change in eGFR over time; hypertension, and 48.0% had diabetes. The mean (SD)
(3) a mixed-effects analysis of eGFR change with a random in- 24-hour urine volume at baseline was 1.9 (0.6) L per day,
tercept to account for center; (4) the annual percentage change eGFR was 43.4 (9.8) mL/min per 1.73 m 2 , and creatinine
([final eGFR−baseline eGFR]÷baseline eGFR); and (5) the pro- clearance was 53.5 (17.4) mL/min per 1.73 m2. Median urine
portion of patients with a 1-year eGFR decline of at least 20%.37 albumin was 122.8 mg/d (IQR, 34.9-512.0). At baseline, 150
To examine the effect of intervention adherence, a prespeci- patients in the control group (47.6% of 315) had a urine vol-
fied per-protocol analysis was conducted that excluded the fol- ume greater than 1.5 L per day (mean, 2.3 L/d) and a urine
lowing: (1) patients in the hydration group who did not main- osmolality of less than 500 mOsm/kg (mean, 365 mOsm/kg);
tain a 24-hour urine volume of at least 0.5 L per day above their these patients were asked to decrease their water intake by
baseline value at 6 and 12 months; (2) patients in the control 1 cup per day.
group who did not maintain a 24-hour urine volume within
0.5 L per day of their baseline value; and (3) patients who Intervention Adherence
missed an assessment or whose final urine sample was col- The mean 1-year change in 24-hour urine volume was 0.6 L per
lected more than 16 months after randomization. Three post day greater in the hydration group than the control group (95%
hoc subgroup analyses were conducted for baseline macroal- CI, 0.5 to 0.7; P < .001), and the mean 9-month change in self-
buminuria, diabetes, and an eGFR below 45 mL/min per reported fluid intake was 0.7 L per day greater in the hydra-
1.73 m2; these analyses were conducted using linear regres- tion group than the control group (95% CI, 0.6 to 0.8; P < .001)
sion with an interaction term. (Table 2).
All analyses except the per-protocol supplementary analy-
sis were conducted according to the intention-to-treat prin- Primary Outcome: Change in eGFR
ciple. All tests were 2-sided and conducted at the .05 level of A scatterplot of the change in eGFR by baseline levels in the hy-
significance. Formal adjustment for multiple comparisons was dration and control groups is shown in eFigure 1 in Supplement
not performed and therefore all analyses of supplementary, sec- 3. The mean 1-year decline in eGFR was −2.2 mL/min per
ondary, and other outcomes should be considered explor- 1.73 m2 in the hydration group and −1.9 mL/min per 1.73 m2 in
atory. Normally distributed variables were compared using the the control group; the adjusted between-group difference in
t test; non-normally distributed variables were compared using change was −0.3 mL/min per 1.73 m2 (95% CI, −1.8 to 1.2; P = .74)
the Wilcoxon 2-sample test using the normal approximation, (Table 3).
and the 95% CI for the difference between 2 medians was cal- Results were similar in the unadjusted analysis and
culated using Hodges-Lehmann estimation. Differences be- when eGFR was calculated using cystatin C (eTable 6 in
tween categorical variables were calculated using the χ2 test, Supplement 3), when eGFR change was analyzed longitudi-
and 95% CIs were calculated with a continuity correction. Sta- nally (eAppendix 2 in Supplement 3), and in a mixed-effects
tistical analyses were conducted using SAS version 9.3 or later, model accounting for the effect of center (eTable 7 in
SPSS version 24 (IBM SPSS), and R (version 3.2.0). Supplement 3). The mean percentage decline in eGFR from
baseline was 4.6% in the hydration group and 4.4% in the
control group (difference, −0.2 [95% CI, −3.8 to 3.5];
P = .93), and the proportion of patients with an eGFR
Results decline of at least 20% over 1 year was 21.6% in the hydra-
Trial enrollment began on April 8, 2013, and the last patient tion group and 20.7% in the control group (difference, 0.9%
was randomized on May 6, 2016 . The sample flow is shown [95% CI, −5.7% to 7.5%]; P = .79) (eTable 8 in Supplement 3).

jama.com (Reprinted) JAMA May 8, 2018 Volume 319, Number 18 1873

© 2018 American Medical Association. All rights reserved.

Downloaded From: on 05/08/2018


Research Original Investigation Effect of Increased Water Intake on Kidney Function Decline in Chronic Kidney Disease

Figure. Flow of Patients Through the Chronic Kidney Disease Water Intake Trial

2390 Individuals approached for


study participationa

1568 Excluded
412 Ineligible
211 Drank more than 10 cups of
water per day
57 Unable to speak or read English
39 Taking a diuretic or lithium
33 Under fluid restriction
72 Other reasonsb
1156 Declined participation
168 Moving, on vacation, or difficulty
getting to laboratories
101 Disliked or unable to drink more water
23 Family situation
864 Other reasonsc

822 Consented

191 Excluded before randomization


112 Ineligible to participate
47 Urine sample >3 L/d
31 No urine sample provided
34 Other reasonsd
78 Withdrew from study
76 Participant decision
2 Physician decision
1 Died a
Prescreened by a research assistant
for eligibility.
b
Other reasons: no urine protein,
631 Randomized
enrolled in another study, pregnant
or breastfeeding, kidney stones in
the past 5 years, less than 2 years of
316 Randomized to hydration group 315 Randomized to control group
316 Received intervention as 315 Received intervention as life expectancy, and gastrointestinal
randomized randomized issues (eg, inflammatory bowel
disease or Crohn disease).
c
5 Diede 7 Diedf Other reasons: too busy, did not
17 Withdrew 4 Withdrew (participant decision) want to undergo blood tests, did
12 Participant decision not want to undergo 24-hour urine
5 Physician decision collections, cancer diagnosis,
physician advised not to participate.
311 Included in primary analysis 308 Included in primary analysis d
Other reasons: medication
5 Excluded (died) 7 Excluded (died) contraindications or chemotherapy.
291 Included in complete case analysis 299 Included in complete case analysis e
Median time to death was 5.1
20 Excluded (missing final eGFR value) 9 Excluded (missing final eGFR value) months (interquartile range, 3.1-7.3).
17 Withdrew from study 4 Withdrew from study (participant f
12 Participant decision decision) Median time to death was 7.2
5 Physician decision 4 Did not complete blood test months (interquartile range,
3 Did not complete blood test for eGFR 2.0-10.2).
for eGFR 1 Started dialysis
eGFR indicates estimated glomerular
filtration rate.

Per-Protocol Analysis Subgroup Analyses


Of 316 patients in the hydration group, 89 (28.2%) main- No statistically significant interactions were observed in 3 post
tained a urine volume that was at least 0.5 L per day greater hoc subgroup analyses of patients with and without macroal-
than their baseline value at each follow-up assessment. Of 315 buminuria (eTable 9 in Supplement 3) or diabetes (eTable 10
patients in the control group, 184 (58.4%) had a urine volume in Supplement 3), and in those with an eGFR of at least
that remained at least 0.5 L per day less than their baseline 45 mL/min per 1.73 m2 or less than 45 mL/min per 1.73 m2 at
value at each follow-up assessment. In these per-protocol baseline (eTable 11 in Supplement 3).
groups, the 1-year change in 24-hour urine volume was 1.5 L
per day more in the hydration group than in the control group Secondary Outcomes
(95% CI, 1.3 to 1.6; P < .001), and the mean 1-year change in As shown in Table 4, statistically significant between-group
eGFR was −1.5 mL/min per 1.73 m2 in the hydration group vs differences were seen for plasma copeptin concentration and
−1.6 mL/min per 1.73 m2 in the control group; the between- creatinine clearance but not for 24-hour urine albumin
group difference was 0.2 mL/min per 1.73 m2 (95% CI, −1.9 to or patient-reported quality of health. The 1-year change in
2.3; P = .86). plasma copeptin concentration was 2.2 pmol/L lower in the

1874 JAMA May 8, 2018 Volume 319, Number 18 (Reprinted) jama.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: on 05/08/2018


Effect of Increased Water Intake on Kidney Function Decline in Chronic Kidney Disease Original Investigation Research

Table 1. Baseline Characteristics of Patients in the Chronic Kidney Disease Table 1. Baseline Characteristics of Patients in the Chronic Kidney Disease
Water Intake Trial (N = 631) Water Intake Trial (N = 631) (continued)

Treatment Group Treatment Group


Demographic and Hydration Control Demographic and Hydration Control
Clinical Characteristics (n = 316) (n = 315) Clinical Characteristics (n = 316) (n = 315)
Age, mean (SD), y 64.8 (12.0) 65.2 (11.6) Measures from blood samples,
mean (SD)
Men, No. (%) 200 (63.3) 200 (63.5)
eGFR (creatinine)c, mL/min 43.2 (10.2) 43.7 (9.3)
Race/ethnicity, No. (%)a per 1.73 m2
White 279 (88.3) 293 (93.0) eGFR (cystatin C)d, mL/min 46.7 (19.8) 47.5 (19.3)
per 1.73 m2
South Asian 14 (4.4) 3 (1.0)
Copeptin, pmol/L 16.6 (11.8) 17.8 (13.7)
Black 7 (2.2) 8 (2.5)
Cystatin C, mg/L 1.6 (0.5) 1.6 (0.4)
Other 16 (5.1) 11 (3.5)
Sodium, mEq/L 139.1 (2.7) 139.2 (2.6)
Weight, mean (SD), kg 87.2 (18.9) 87.2 (19.9)
Osmolality, mOsm/kg 300.3 (7.4) 301.2 (8.3)
BMI,b mean (SD) 30.0 (6.0) 29.8 (6.0)
b Urea, mg/dL 28.9 (9.5) 28.6 (10.4)
Obese (BMI ≥30 ), No. (%) 149 (47.2) 139 (44.1)
Measures from 24-h
Current smoker, No. (%) 30 (9.5) 38 (12.1) urine samples
Daily fluid intake, 2.1 (0.8) 2.0 (0.7) Urine volume, mean (SD), L/d 1.9 (0.6) 1.9 (0.6)
mean (SD), L/d
Creatinine, mean (SD), mmol/d 11.9 (4.3) 12.0 (3.7)
Primary etiology
of chronic kidney disease, Osmolality, mean (SD), mOsm/kg 447.4 (140.0) 456.6 (132.9)
No. (%)
Urea, mean (SD), mmol/d 339.6 (129.9) 350.2 (125.7)
Hypertension 127 (40.2) 119 (37.8)
Sodium, mean (SD), mmol/d 145.9 (62.4) 149.2 (62.5)
Diabetes 120 (38.0) 105 (33.3)
Potassium, mean (SD), mmol/d 63.3 (26.6) 63.9 (23.4)
Glomerulonephritis 35 (11.1) 41 (13.0)
Albumin, median (IQR), mg/d 108 (31-522) 150 (40-507)
Autoimmune 15 (4.7) 26 (8.3)
Creatinine clearance, mean (SD), 53.0 (18.7) 54.0 (16.4)
Polycystic kidney disease 9 (2.9) 9 (2.8) mL/min per 1.73 m2
Other/unknown 10 (3.2) 15 (4.8) Medications, No. (%)
Chronic kidney disease ACE inhibitor or angiotensin 217 (68.7) 211 (67.0)
stage, No. (%) receptor blocker
c
eGFR ≥45 mL/min 141 (44.6) 148 (47.0) Statin 203 (64.2) 211 (67.0)
per 1.73 m2
Calcium channel blockers 134 (42.4) 125 (39.7)
Urine albumin <30 mg/d 28 (8.9) 39 (12.4)
β-Blockers 124 (39.2) 127 (40.3)
Urine albumin 30-300 mg/d 58 (18.4) 65 (20.6)
Diuretic 133 (42.1) 113 (35.9)
Urine albumin >300 mg/d 49 (15.5) 33 (10.5)
eGFRc<45 mL/min 175 (55.4) 167 (53.0) Abbreviations: ACE, angiotensin-converting enzyme; BMI, body mass index;
per 1.73 m2 CKD-EPI, Chronic Kidney Disease Epidemiology Collaboration; eGFR, estimated
glomerular filtration rate; IQR, interquartile range.
Urine albumin <30 mg/d 31 (9.8) 33 (10.5)
SI conversion factors: To convert serum urea to mmol/L, multiply by 0.357.
Urine albumin 30-300 mg/d 77 (24.4) 63 (20.0)
a
Self-reported race/ethnicity was collected and recorded by a research
Urine albumin >300 mg/d 53 (16.8) 63 (20.0)
assistant using prespecified categories; black/white racial origin is needed for
Comorbidities within the calculation of eGFR.
last 10 y, No. (%) b
BMI was calculated as weight in kilograms divided by height in meters squared.
Hypertension 278 (88.3) 278 (88.3) c
Calculated using the CKD-EPI creatinine equation.
Hyperlipidemia 211 (67.2) 206 (65.8) d
Calculated using the CKD-EPI cystatin C equation.
Diabetes 166 (52.7) 137 (43.5)
Malignancy 60 (19.1) 72 (23.0)
hydration group than the control group (95% CI, −3.9 to −0.5;
Coronary artery disease 64 (20.4) 67 (21.3)
P = .01), and the 1-year change in creatinine clearance was
Cerebrovascular/transient 36 (11.5) 45 (14.3)
ischemic attack 3.6 mL/min per 1.73 m2 higher in the hydration group than the
Peripheral vascular disease 28 (8.9) 28 (8.9) control group (95% CI, 0.8 to 6.4; P = .01). The between-
Chronic obstructive 45 (14.2) 43 (13.7) group difference in the 1-year change in urine albumin was
pulmonary disease 6.8 mg/d (95% CI, −3.9 to 50.8; P = .11), and the between-
Transplant 13 (4.1) 20 (6.3) group difference in the 1-year change in quality of life was 0.2
Congestive heart failure 19 (6.0) 11 (3.5) units (95% CI, −0.3 to 0.3; P = .22).
Family history of hypertension 181 (57.3) 177 (56.2)
or kidney failure, No. (%)
Systolic blood pressure, 140.8 (19.1) 136.8 (17.2)
Other Outcomes
mean (SD), mm Hg The mean 1-year change in 24-hour urine osmolality, creati-
Diastolic blood pressure, 79.2 (10.3) 77.7 (10.9) nine, urea, sodium, and potassium are shown in eTable 12 in
mean (SD), mm Hg
Supplement 3; between-group differences were −75.6 mOsm/kg
(continued) (95% CI, −98.2 to −53.0; P < .001) for urine osmolality,
0.8 mmol per day (95% CI, 0.3 to 1.4; P = .003) for creatinine,

jama.com (Reprinted) JAMA May 8, 2018 Volume 319, Number 18 1875

© 2018 American Medical Association. All rights reserved.

Downloaded From: on 05/08/2018


Research Original Investigation Effect of Increased Water Intake on Kidney Function Decline in Chronic Kidney Disease

Table 2. Change in 24-Hour Urine Volume and Self-reported Fluid Intakea

Mean (SD)
Hydration Group Control Group Mean Difference
(n = 291) (n = 299) (95% CI)b P Valueb
Urine volume, L/d a
All analyses followed the
Prerandomization 1.9 (0.6) 1.9 (0.6) intention-to-treat principle.
b
6 Month 2.5 (0.9) 1.9 (0.6) Between-group differences in
change were analyzed using the
12 Month 2.5 (0.8) 1.9 (0.7)
independent-samples t test.
12-Month change (95% CI) 0.6 (0.5 to 0.7)c −0.04 (0.0 to 0.1)c 0.6 (0.5 to 0.7) <.001 c
Change was calculated as the final
Self-reported fluid intake, L/d value minus the prerandomization
Prerandomization 2.1 (0.8) 2.0 (0.7) value; restricted to patients who
provided follow-up data within 8 to
3 Month 2.8 (0.9) 2.0 (0.6)
16 months of randomization
6 Month 2.8 (0.8) 2.1 (0.7) (excluding 12 patients who died
9 Month 2.8 (0.8) 2.0 (0.6) during the 12-month trial period
[5 in the hydration group and 7 in
9-Month change (95% CI) 0.7 (0.6 to 0.8)c 0.0 (−0.1 to 0.1)c 0.7 (0.6 to 0.8) <.001
the control group]).

Table 3. Primary Outcome: 1-Year Change in Estimated Glomerular Filtration Ratea

Mean (95% CI)


Hydration Group Control Group Adjusted Between-Group Difference
eGFR, mL/min per 1.73 m2 (n = 311) (n = 308) in Changeb (95% CI) P Value
Prerandomization 43.3 (42.1 to 44.4) 43.6 (42.6 to 44.7)
12 Months 41.0 (39.5 to 42.6) 41.7 (40.3 to 43.1)
Change −2.2 (−3.3 to −1.1)c −1.9 (−2.9 to −0.9)c −0.3 (−1.8 to 1.2) .74
Abbreviation: eGFR, estimated glomerular filtration rate. current smoker, presence of diabetes, urinary albumin (mg/d)
a
Analyses exclude 12 patients who died during the 12-month trial period (log-transformed), and use of the following medications: an angiotensin-
(5 in the hydration group; 7 in the control group); all analyses followed the converting enzyme inhibitor or angiotensin receptor blocker, diuretic,
intention-to-treat principle. Fully conditionally specified models were used to β-blocker, calcium channel blocker, and statin.
c
impute missing 12-month eGFR values for 29 patients. The mean change in eGFR (12-month value minus the prerandomization value)
b
The adjusted between-group difference in eGFR change was analyzed using was calculated within each of 20 imputed data sets, and the mean was
linear regression, adjusted for age (in years), sex, obesity (body mass index calculated using the imputed data set means.
ⱖ30 [calculated as weight in kilograms divided by height in meters squared]),

25.4 mmol per day (95% CI, 5.1 to 45.6; P = .01) for urea, each case was investigated and reported to the participant’s
21.9 mmol per day (95% CI, 10.3 to 33.6; P < .001) for sodium, physician; all values normalized by the next study visit.
and 2.8 mmol per day (95% CI, −1.6 to 7.1; P = .22) for potas- The 1-year change in serum sodium was 0.5 mEq/L lower
sium. No statistically significant differences were seen be- in the hydration group than the control group (95% CI, −1.0
tween groups for the 1-year change in serum osmolality, to −0.04; P = .04).
creatinine, or urea (eTable 13 in Supplement 3); blood pres-
sure, weight, waist circumference, or body mass index (eTable
14 in Supplement 3); or the 6-month change in dietary intake
of protein and sodium (eTable 15 in Supplement 3).
Discussion
In this randomized clinical trial of approximately 600 pa-
Adverse Events and Serum Sodium Monitoring tients with stage 3 chronic kidney disease, drinking more wa-
The data and safety monitoring board performed 5 reviews ter (compared with usual fluid intake alone) did not slow the
of trial safety and integrity during the recruitment period decline in eGFR over 1 year. This trial had 80% statistical power
and recommended that the trial continue. During the trial, to detect a between-group difference in eGFR change of
52 adverse events occurred among 42 (13%) patients in the 2 mL/min per 1.73 m2. Findings from the primary intention-
hydration group (including 5 deaths [3 additional deaths to-treat analyses were consistent with the per-protocol analy-
occurred between 13 and 16 months]) and 66 events ses for which there was a greater separation in 24-hour urine
occurred among 51 (16%) patients in the control group volume between groups.
(including 7 deaths [3 additional deaths occurred between The absence of an effect of drinking more water on eGFR
13 and 16 months]). All adverse events were investigated, may be interpreted in several ways. First, increased water
and no events were attributed to the intervention. Patients’ intake may not protect against declining kidney function,
serum sodium levels were monitored also at 3-month inter- and the results of prior observational studies may have
vals (eTable 16 in Supplement 3). Serum sodium concentra- been confounded.9-11
tion declined to less than 130 mEq/L in 3 patients in the Second, there may have been an effect, but the study
hydration group and in 1 participant in the control group; was underpowered to identify it as statistically significant

1876 JAMA May 8, 2018 Volume 319, Number 18 (Reprinted) jama.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: on 05/08/2018


Effect of Increased Water Intake on Kidney Function Decline in Chronic Kidney Disease Original Investigation Research

Table 4. Secondary Outcomes: 1-Year Change in Plasma Copeptin, Creatinine Clearance, 24-Hour Urine Albumin,
and Patient-Reported Overall Quality of Healtha
Hydration Group Control Group Between-Group
(n = 291) (n = 299) Difference in Changeb (95% CI) P Valueb
Plasma copeptin, mean (SD), pmol/L
Prerandomization 17.4 (12.4) 17.5 (13.0)
12 Month 16.1 (13.5) 18.3 (13.6)
Change (95% CI) −1.4 (−2.7 to −0.0)c 0.8 (−0.3 to 1.9)c −2.2 (−3.9 to −0.5) .01
Creatinine clearance, mean (SD), mL/min
per 1.73 m2
Prerandomization 52.5 (17.1) 54.6 (15.6)
12 Month 53.1 (19.7) 51.6 (18.9)
Change (95% CI) 0.6 (−1.6 to 2.7)c −3.0 (−4.9 to −1.2)c 3.6 (0.8 to 6.4) .01
Urine albumin, mean (SD), mg/d
Prerandomization 139 (49 to 425) 108 (32 to 606)
12 Month 142 (38 to 509) 103 (32 to 497)
Change (95% CI) 7 (−4.1 to 16.2)c 0.2 (−7.0 to 4.6)c 6.8 (−3.9 to 50.8) .11
Quality of healthd
Prerandomization 7.3 (1.6) 7.3 (1.6)
12 Month 7.2 (1.9) 7.1 (1.8)
Change (95% CI) −0.0 (−0.3 to 0.2)c −0.2 (−0.4 to −0.0)c 0.2 (−0.3 to 0.3) .22
Abbreviation: IQR, interquartile range. data); CIs for the difference between 2 medians were calculated using
a
All analyses followed the intention-to-treat principle. No adjustment was Hodges-Lehmann estimation.
c
made for multiple comparisons; therefore all results should be interpreted Change was calculated as the 12-month value minus the prerandomization
as exploratory. value; restricted to patients who provided follow-up data within 8 to 16
b
Between-group differences in change were analyzed using the months of randomization (excluding 12 patients who died during the 12-mo
independent-samples t test (for normally distributed data) or the Wilcoxon trial period [5 in the hydration group and 7 in the control group]).
d
2-sample test using the normal approximation (for non-normally distributed Measured on a 10-point Likert scale (0 [worst possible] to 10 [best possible]).

(this study was underpowered to detect a between-group serum cystatin C was −0.2 mL/min per 1.73 m2 (P > .05); how-
difference less than 2 mL/min per 1.73 m2); however, the 1-year ever, the 1-year change in creatinine clearance was signifi-
decline in eGFR was slightly greater in the hydration group, cantly higher in the hydration group relative to the control
suggesting a possible effect in the opposite direction to that group (the between-group difference was 3.5 mL/min per
originally hypothesized. 1.73 m2 [P = .01]). The latter finding appeared to be driven
Third, although the hydration group achieved a signifi- by an increased urinary excretion of creatinine in the hydra-
cantly increased fluid intake and decreased copeptin concen- tion group relative to the control group (eTable 12 in Supple-
trations relative to the control group, perhaps a greater differ- ment 3). Three explanations for this finding were considered.
ence in water intake is needed to produce a measurable effect Given that the urinary excretion of urea and sodium were
on eGFR (although the per-protocol analysis was consistent also significantly increased in the hydration group relative to
with the intent-to-treat analysis). the control group (eTable 12 in Supplement 3), the possibility
Fourth, more than 1 year of follow-up may be needed to of systematic urine collection errors was considered; how-
detect an effect on eGFR. In a clinical trial of patients with ever, urine osmolality and plasma copeptin both decreased
autosomal dominant polycystic kidney disease treated with significantly more in the hydration group relative to the con-
Tolvaptan (a vasopressin V2-receptor antagonist) vs placebo, trol group, suggesting that patients were following their
between-group differences in kidney function did not randomly assigned intervention. In addition, an increased
emerge until 24 to 36 months after randomization.38 Simi- urinary excretion of creatinine, urea, and sodium may reflect
larly, a 7-year observational study of 2000 healthy adults an increase in weight (eTable 14 in Supplement 3) or intake of
showed less eGFR decline with larger urine outputs. 9 protein and sodium (eTable 15 in Supplement 3); however, no
Although patients in the present trial will undergo an addi- between-group differences in these variables were observed,
tional 12 months of follow-up to evaluate 2-year eGFR although perhaps there were dietary changes that went unre-
change, hydration coaching stopped at 12 months, which corded. Also, it is possible that patients in the hydration
may negatively affect patients’ adherence to their random group had an increased urinary flow rate, which has been
allocation.16 shown to cause increased tubular secretion and decreased
A marked incongruence was observed between the 1-year reabsorption of urinary solutes (specifically creatinine and
change in eGFR and creatinine clearance. The between- urea).39,40 The clinical significance of an increased urinary
group difference in the 1-year change in GFR estimated flow rate is unknown and further research is needed to better
with serum creatinine was −0.3 mL/min per 1.73 m2 and with understand this finding and the underlying physiology.

jama.com (Reprinted) JAMA May 8, 2018 Volume 319, Number 18 1877

© 2018 American Medical Association. All rights reserved.

Downloaded From: on 05/08/2018


Research Original Investigation Effect of Increased Water Intake on Kidney Function Decline in Chronic Kidney Disease

Limitations Third, the generalizability of trial findings may be limited


This study has several limitations. First, despite the efforts of by enrollment restricted to patients in Southwestern Ontario,
dieticians and research assistants who coached patients in the Canada (a northern region with a temperate climate) in which
hydration group to drink 1.0 to 1.5 L of water per day (depend- 90% of the patients were white.
ing on sex and weight), mean urine volume in the hydration Fourth, the use of estimated rather than measured GFR
group increased by only 0.6 L per day (2.4 cups) relative to the represents an important source of measurement error in this
control group. This highlights how difficult it would be to study. Exogenously measured GFR may be more sensitive to
achieve a large sustained increase in water intake in routine changes in kidney function; however, measurement proto-
practice. However, the increased water intake achieved in this cols are more invasive and costly, which may be considered
trial was sufficient to lower vasopressin secretion, as as- unacceptable to patients.
sessed by plasma copeptin concentrations (the 1-year change
in plasma copeptin was 2.3 pmol/L lower in the hydration group
than the control group [P = .009]).
Second, although the magnitude of eGFR decline was
Conclusions
0.3 mL/min per 1.73 m2 greater in the hydration group rela- Among adults with stage 3 chronic kidney disease, coaching
tive to the control group, the 95% CI around this estimate to increase water intake compared with coaching to main-
spanned −1.8 to 1.2, and the study was underpowered to de- tain the same water intake did not significantly slow the
tect the prespecified minimal clinically important difference decline in kidney function after 1 year. However, the study
of 1 mL/min per 1.73 m2; therefore, trial findings should be in- may have been underpowered to detect a clinically impor-
terpreted as null but potentially underpowered. tant difference.

ARTICLE INFORMATION Role of Funder/Sponsor: Danone Research failure in the 5/6 nephrectomized rat. Am J Physiol.
Accepted for Publication: March 29, 2018. requested that this study include the measure of 1990;258(4 Pt 2):F973-F979.
creatinine clearance as a secondary outcome; aside 9. Clark WF, Sontrop JM, Macnab JJ, et al. Urine
Author Contributions: Drs Clark and Sontrop had from this, the study sponsors had no role in the
full access to all the data in the study and take volume and change in estimated GFR in a
design and conduct of the study; collection, community-based cohort study. Clin J Am Soc
responsibility for the integrity of the data and the management, analysis, and interpretation of the
accuracy of the data analysis. Nephrol. 2011;6(11):2634-2641.
data; preparation, review, or approval of the
Concept and design: Clark, Sontrop, Huang, Gallo, manuscript; and decision to submit the manuscript 10. Sontrop JM, Dixon SN, Garg AX, et al.
Moist, House, Weir, Garg. for publication. Association between water intake, chronic
Acquisition, analysis, or interpretation of data: Clark, kidney disease, and cardiovascular disease:
Sontrop, Huang, Gallo, House, Cuerden, Weir, Additional Contributions: Michael Walsh, MD, PhD a cross-sectional analysis of NHANES data. Am J
Bagga, Brimble, Burke, Muirhead, Pandeya, Garg. (McMaster University), and Daniel G. Biche, MD Nephrol. 2013;37(5):434-442.
Drafting of the manuscript: Clark, Sontrop, (University of Montreal), served on the data and
safety monitoring board for this trial. Ivan Tack, MD, 11. Strippoli GF, Craig JC, Rochtchina E, Flood VM,
Huang, Garg. Wang JJ, Mitchell P. Fluid and nutrient intake and
Critical revision of the manuscript for important PhD (Toulouse Hospital), provided useful insight on
renal physiology. None of these individuals received risk of chronic kidney disease. Nephrology (Carlton).
intellectual content: All authors. 2011;16(3):326-334.
Statistical analysis: Sontrop, Cuerden, Garg. compensation for their contribution. We also
Obtained funding: Clark, Sontrop. acknowledge the 631 patients without whom this 12. Qaseem A, Dallas P, Forciea MA, Starkey M,
Administrative, technical, or material support: Clark, investigation would not be possible. Denberg TD; Clinical Guidelines Committee of the
Huang, Gallo, Moist, Weir, Bagga, Burke, American College of Physicians. Dietary and
Muirhead, Garg. REFERENCES pharmacologic management to prevent recurrent
Supervision: Clark, Huang, Gallo, Moist, Weir, 1. Food and Nutrition Board—National Academy of nephrolithiasis in adults: a clinical practice guideline
Brimble, Muirhead, Pandeya. Sciences. Recommended Dietary Allowances. Natl from the American College of Physicians. Ann Intern
Res Counc. 1945;No. 122:3-18. Med. 2014;161(9):659-667.
Conflict of Interest Disclosures: All authors have
completed and submitted the ICMJE Form for 2. Valtin H. “Drink at least eight glasses of water 13. Borghi L, Meschi T, Amato F, Briganti A, Novarini
Disclosure of Potential Conflicts of Interest. Dr Clark a day.” really? is there scientific evidence for “8 x 8”? A, Giannini A. Urinary volume, water and
reports receipt of an unrestricted grant, personal Am J Physiol Regul Integr Comp Physiol. 2002;283 recurrences in idiopathic calcium nephrolithiasis:
fees (scientific advisory board), and travel support (5):R993-R1004. a 5-year randomized prospective study. J Urol.
from Danone Research. Dr Sontrop reports other 1996;155(3):839-843.
3. Vreeman RC, Carroll AE. Medical myths. BMJ.
(travel support) from Danone Research. Dr Huang 2007;335(7633):1288-1289. 14. Clark WF, Sontrop JM, Huang SH, et al.
reports receipt of a grant from Danone Research. The chronic kidney disease Water Intake Trial (WIT):
Dr Moist reports other (travel support and serving 4. Lette F, Dwyer JP. The fluid craze. Lancet. 2008; results from the pilot randomised controlled trial.
as chair of the ISN/Danone Hydration for Kidney 372(9641):782-784. BMJ Open. 2013;3(12):e003666.
Health Research Initiative) from Danone Nutrica. 5. Clark WF, Sontrop JM, Huang S-H, Moist L, 15. Sontrop JM, Huang S-H, Garg AX, et al. Effect of
No other disclosures were reported. Bouby N, Bankir L. Hydration and chronic kidney increased water intake on plasma copeptin in
Funding/Support: This study was funded by disease progression: a critical review of the patients with chronic kidney disease: results from a
Danone Research and the Program of Experimental evidence. Am J Nephrol. 2016;43(4):281-292. pilot randomised controlled trial. BMJ Open. 2015;
Medicine, Western University, Canada. Thermo 6. Wang CJ, Grantham JJ, Wetmore JB. 5(11):e008634.
Fisher Scientific provided the instrumentation, The medicinal use of water in renal disease. Kidney 16. Clark WF, Huang S-HS, Garg AX, et al.
assay reagent, and disposables for the analysis Int. 2013;84(1):45-53. The Chronic Kidney Disease Water Intake Trial
copeptin. Dr Garg was supported by the Dr Adam 7. Bankir L, Bouby N, Ritz E. Vasopressin: a novel (WIT): protocol of a randomized controlled
Linton Chair in Kidney Health Analytics, and a target for the prevention and retardation of kidney trial[published online August 22, 2017]. Can J
Clinician Salary Award from the Canadian Institutes disease? Nat Rev Nephrol. 2013;9(4):223-239. Kidney Heal Dis. doi:10.1177/2054358117725106
of Health Research.
8. Bouby N, Bachmann S, Bichet D, Bankir L. Effect 17. Boutron I, Moher D, Altman DG, Schulz KF,
of water intake on the progression of chronic renal Ravaud P; CONSORT Group. Extending the

1878 JAMA May 8, 2018 Volume 319, Number 18 (Reprinted) jama.com

© 2018 American Medical Association. All rights reserved.

Downloaded From: on 05/08/2018


Effect of Increased Water Intake on Kidney Function Decline in Chronic Kidney Disease Original Investigation Research

CONSORT statement to randomized trials of version 1.3. 1997. http://www.rand.org/pubs/papers 33. Lorenzo V, Saracho R, Zamora J, Rufino M,
nonpharmacologic treatment: explanation and /P7994.html. Accessed January 12, 2017. Torres A. Similar renal decline in diabetic and
elaboration. Ann Intern Med. 2008;148(4):295-309. 25. Hays RD, Kallich JD, Mapes DL, Coons SJ, non-diabetic patients with comparable levels of
18. National Kidney Foundation. KDOQI clinical Carter WB. Development of the kidney disease albuminuria. Nephrol Dial Transplant. 2010;25(3):
practice guidelines for chronic kidney disease: quality of life (KDQOL) instrument. Qual Life Res. 835-841.
evaluation, classification, and stratification. Am J 1994;3(5):329-338. 34. Moher D, Hopewell S, Schulz KF, et al.
Kidney Dis. 2002;39(2 Suppl 1):S1-S266. 26. Matsushita K, Selvin E, Bash LD, Franceschini CONSORT 2010 explanation and elaboration:
19. Wang CJ, Creed C, Winklhofer FT, Grantham JJ. N, Astor BC, Coresh J. Change in estimated GFR updated guidelines for reporting parallel group
Water prescription in autosomal dominant associates with coronary heart disease and randomised trials. BMJ. 2010;340:c869.
polycystic kidney disease: a pilot study. Clin J Am mortality. J Am Soc Nephrol. 2009;20(12):2617-2624. 35. Roy J, Shou H, Xie D, et al; Chronic Renal
Soc Nephrol. 2011;6(1):192-197. 27. Turin TC, Coresh J, Tonelli M, et al. One-year Insufficiency Cohort (CRIC) Study Investigators.
20. Barash I, Ponda MP, Goldfarb DS, Skolnik EY. change in kidney function is associated with an Statistical methods for cohort studies of CKD:
A pilot clinical study to evaluate changes in urine increased mortality risk. Am J Nephrol. 2012;36(1): prediction modeling. Clin J Am Soc Nephrol. 2017;12
osmolality and urine cAMP in response to acute and 41-49. (6):1010-1017.
chronic water loading in autosomal dominant 28. Roussel R, Fezeu L, Marre M, et al. Comparison 36. Van Buuren S, Brand JPL, Groothuis-Oudshoorn
polycystic kidney disease. Clin J Am Soc Nephrol. between copeptin and vasopressin in a population CGM, Rubin DB. Fully conditional specification
2010;5(4):693-697. from the community and in people with chronic in multivariate imputation. J Stat Comput Simul.
21. Spigt MG, Knottnerus JA, Westerterp KR, Olde kidney disease. J Clin Endocrinol Metab. 2014;99 2006;76(12):1049-1064. doi:10.1080
Rikkert MG, Schayck CP. The effects of 6 months (12):4656-4663. /10629360600810434.
of increased water intake on blood sodium, 29. Enhörning S, Christensson A, Melander O. 37. Coresh J, Turin TC, Matsushita K, et al. Decline
glomerular filtration rate, blood pressure, and Plasma copeptin as a predictor of kidney in estimated glomerular filtration rate and
quality of life in elderly (aged 55-75) men. J Am disease[published online February 16, 2018]. subsequent risk of end-stage renal disease and
Geriatr Soc. 2006;54(3):438-443. Nephrol Dial Transplant. doi:10.1093/ndt/gfy017. mortality. JAMA. 2014;311(24):2518-2531.
22. Levey AS, Stevens LA, Schmid CH, et al; 30. Cravedi P, Ruggenenti P, Remuzzi G. 38. Torres VE, Chapman AB, Devuyst O, et al;
CKD-EPI (Chronic Kidney Disease Epidemiology Proteinuria should be used as a surrogate in CKD. TEMPO 3:4 Trial Investigators. Tolvaptan in patients
Collaboration). A new equation to estimate Nat Rev Nephrol. 2012;8(5):301-306. with autosomal dominant polycystic kidney
glomerular filtration rate. Ann Intern Med. 2009; disease. N Engl J Med. 2012;367(25):2407-2418.
150(9):604-612. 31. Rosansky SJ. Renal function trajectory is more
important than chronic kidney disease stage for 39. Sjöström PA, Odlind BG, Wolgast M. Extensive
23. Christ-Crain M, Fenske W. Copeptin in the managing patients with chronic kidney disease. Am tubular secretion and reabsorption of creatinine in
diagnosis of vasopressin-dependent disorders of J Nephrol. 2012;36(1):1-10. humans. Scand J Urol Nephrol. 1988;22(2):129-131.
fluid homeostasis. Nat Rev Endocrinol. 2016;12(3): 40. Joppich R, Deetjen P. The relation between the
168-176. 32. Eriksen BO, Tomtum J, Ingebretsen OC.
Predictors of declining glomerular filtration rate in reabsorption of urea and of water in the distal
24. Hays RD, Kallich J, Mapes D, et al. Kidney a population-based chronic kidney disease cohort. tubule of the rat kidney. Pflugers Arch. 1971;329(2):
Disease Quality of Life Short Form (KDQOL-SF TM), Nephron Clin Pract. 2010;115(1):c41-c50. 172-185.

jama.com (Reprinted) JAMA May 8, 2018 Volume 319, Number 18 1879

© 2018 American Medical Association. All rights reserved.

Downloaded From: on 05/08/2018

Potrebbero piacerti anche