ArticlePDF Available

The effect of CCR2 inhibitor CCX140-B on residual albuminuria in patients with type 2 diabetes and nephropathy: A randomised trial

Authors:

Abstract and Figures

Patients with type 2 diabetes and nephropathy have high cardiorenal morbidity and mortality despite optimum treatment including angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs). Residual risk is related to residual albuminuria. We assessed whether CCX140-B, a selective inhibitor of C-C chemokine receptor type 2 (CCR2), could further reduce albuminuria when given in addition to standard care, including ACE inhibitors or ARBs. In this randomised, double-blind, placebo-controlled clinical trial, we recruited patients from 78 research centres in Belgium, Czech Republic, Germany, Hungary, Poland, and the UK. We enrolled patients with type 2 diabetes aged 18-75 years with proteinuria (first morning void urinary albumin to creatinine ratio [UACR] 100-3000 mg/g), estimated glomerular filtration rate of 25 mL/min per 1·73 m(2) or higher, and taking stable antidiabetic treatment and ACE inhibitors or ARBs, for at least 8 weeks before study entry. Patients were stratified based on baseline UACR and renal function (estimated glomerular filtration rate), and then randomly assigned (1:1:1) via an interactive web response system with a minimisation algorithm to oral placebo, 5 mg CCX140-B, or 10 mg CCX140-B once a day. The 12-week dosing period in the initial protocol was extended to 52 weeks by protocol amendment. The primary efficacy measure was change from baseline in UACR during 52 weeks in the modified intention-to-treat population (all patients with uninterrupted dosing, excluding patients who stopped dosing at week 12 either permanently under the original protocol, or temporarily because of delay in approval of the protocol amendment). We did safety analyses on all randomly assigned patients who received at least one dose of study drug. According to a prespecified analysis plan, we analysed the primary endpoint with one-sided statistical testing with calculation of upper 95% confidence limits of the differences between active and control. This trial is registered with ClinicalTrials.gov, number NCT01447147. The study ran from Dec 7, 2011 (first patient enrolled), until Aug 4, 2014. We enrolled 332 patients: 111 were assigned to receive placebo, 110 to 5 mg CCX140-B, and 111 to 10 mg CCX140-B. Of these, 192 were included in the modified intention-to-treat population. UACR changes from baseline during 52 weeks were -2% for placebo (95% CI -11% to 9%), -18% for 5 mg CCX140-B (-26% to -8%), and -11% for 10 mg CCX140-B (-20% to -1%). We recorded a -16% difference between 5 mg CCX140-B and placebo (one-sided upper 95% confidence limit -5%; p=0·01) and a -10% difference between 10 mg CCX140-B and placebo (upper 95% confidence limit 2%; p=0·08). Adverse events occurred in 81 (73%) of 111 patients in the placebo group versus 71 (65%) of 110 patients in the CCX140-B 5 mg group and 68 (61%) of 111 patients in the CCX140-B 10 mg group; there were no renal events during the study. Our data suggest that CCR2 inhibition with CCX140-B has renoprotective effects on top of current standard of care in patients with type 2 diabetes and nephropathy. ChemoCentryx. Copyright © 2015 Elsevier Ltd. All rights reserved.
Content may be subject to copyright.
www.thelancet.com/diabetes-endocrinology Published online August 10, 2015 http://dx.doi.org/10.1016/S2213-8587(15)00261-2
1
Articles
The eff ect of CCR2 inhibitor CCX140-B on residual
albuminuria in patients with type 2 diabetes and
nephropathy: a randomised trial
Dick de Zeeuw, Pirow Bekker, Elena Henkel, Christopher Hasslacher, Ioanna Gouni-Berthold, Heidrun Mehling, Antonia Potarca, Vladimir Tesar,
Hiddo J Lambers Heerspink, Thomas J Schall, for the CCX140-B Diabetic Nephropathy Study Group*
Summary
Background Patients with type 2 diabetes and nephropathy have high cardiorenal morbidity and mortality despite
optimum treatment including angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers
(ARBs). Residual risk is related to residual albuminuria. We assessed whether CCX140-B, a selective inhibitor of C-C
chemokine receptor type 2 (CCR2), could further reduce albuminuria when given in addition to standard care,
including ACE inhibitors or ARBs.
Methods In this randomised, double-blind, placebo-controlled clinical trial, we recruited patients from 78 research
centres in Belgium, Czech Republic, Germany, Hungary, Poland, and the UK. We enrolled patients with type 2
diabetes aged 18–75 years with proteinuria (fi rst morning void urinary albumin to creatinine ratio [UACR]
100–3000 mg/g), estimated glomerular fi ltration rate of 25 mL/min per 1·73 m² or higher, and taking stable
antidiabetic treatment and ACE inhibitors or ARBs, for at least 8 weeks before study entry. Patients were stratifi ed
based on baseline UACR and renal function (estimated glomerular fi ltration rate), and then randomly assigned (1:1:1)
via an interactive web response system with a minimisation algorithm to oral placebo, 5 mg CCX140-B, or 10 mg
CCX140-B once a day. The 12-week dosing period in the initial protocol was extended to 52 weeks by protocol
amendment. The primary effi cacy measure was change from baseline in UACR during 52 weeks in the modifi ed
intention-to-treat population (all patients with uninterrupted dosing, excluding patients who stopped dosing at
week 12 either permanently under the original protocol, or temporarily because of delay in approval of the protocol
amendment). We did safety analyses on all randomly assigned patients who received at least one dose of study drug.
According to a prespecifi ed analysis plan, we analysed the primary endpoint with one-sided statistical testing with
calculation of upper 95% confi dence limits of the diff erences between active and control. This trial is registered with
ClinicalTrials.gov, number NCT01447147.
Findings The study ran from Dec 7, 2011 (fi rst patient enrolled), until Aug 4, 2014. We enrolled 332 patients: 111 were
assigned to receive placebo, 110 to 5 mg CCX140-B, and 111 to 10 mg CCX140-B. Of these, 192 were included in the
modifi ed intention-to-treat population. UACR changes from baseline during 52 weeks were –2% for placebo (95% CI
–11% to 9%), –18% for 5 mg CCX140-B (–26% to –8%), and –11% for 10 mg CCX140-B (–20% to –1%). We recorded
a –16% diff erence between 5 mg CCX140-B and placebo (one-sided upper 95% confi dence limit –5%; p=0·01) and a
–10% diff erence between 10 mg CCX140-B and placebo (upper 95% confi dence limit 2%; p=0·08). Adverse events
occurred in 81 (73%) of 111 patients in the placebo group versus 71 (65%) of 110 patients in the CCX140-B 5 mg group
and 68 (61%) of 111 patients in the CCX140-B 10 mg group; there were no renal events during the study.
Interpretation Our data suggest that CCR2 inhibition with CCX140-B has renoprotective eff ects on top of current
standard of care in patients with type 2 diabetes and nephropathy.
Funding ChemoCentryx.
Introduction
Patients with type 2 diabetes and proteinuria have a
high renal and cardiovascular morbidity and mortality.
Treatment of diabetic nephropathy, including with
angiotensin-converting enzyme (ACE) inhibitors or
angiotensin receptor blockers (ARBs), has been
e ective in reducing cardiovascular and renal risk.
These renal and cardiovascular protective properties of
renin-angiotensin-system (RAS) inhibitors have,
besides blood pressure lowering, been partly attributed
to their albuminuria lowering e ect.1–5 Despite the
success of RAS inhibitors, residual renal and
cardiovascular risk is very high6 and seems to be related
to the residual high albuminuria in these patients.1
Increasing the blockade of the RAS with ACE inhibitors,
ARBs, or renin-inhibition combinations has not been
successful in further reducing renal or cardiovascular
risk in this patient population.7,8
Novel treatment options that target other pathways
involved in the pathophysiology of diabetic nephropathy
are needed. Monocyte chemoattractant protein-1
(MCP-1), also called C-C chemokine ligand 2 (CCL2),
Lancet Diabetes Endocrinol 2015
Published Online
August 10, 2015
http://dx.doi.org/10.1016/
S2213-8587(15)00261-2
See Online/Comment
http://dx.doi.org/10.1016/
S2213-8587(15)00286-7
*Members listed in the appendix
Department of Clinical
Pharmacy and Pharmacology,
University of Groningen,
University Medical Center
Groningen (UMCG), Groningen,
the Netherlands
(Prof D de Zeeuw MD,
H J Lambers Heerspink PharmD);
ChemoCentryx, Mountain
View, CA, USA (P Bekker MBChB,
A Potarca MSc, T J Schall PhD);
Centre for Clinical Studies,
Technical University, Dresden,
Germany (E Henkel MD);
Diabetes Institute, Heidelberg,
Germany (C Hasslacher MD);
Center for Endocrinology,
Diabetes and Preventive
Medicine, University of
Cologne, Cologne, Germany
(I Gouni-Berthold MD); Charité
Experimental and Clinical
Research Centre, Berlin,
Germany (H Mehling MD); and
Department of Nephrology,
1st School of Medicine, Charles
University, Prague, Czech
Republic (Prof V Tesar MD)
Correspondence to:
Prof Dick de Zeeuw, Department
of Clinical Pharmacy and
Pharmacology, University
Medical Center Groningen,
9700 AD Groningen,
Netherlands
d.de.zeeuw@umcg.nl
See Online for appendix
Articles
2
www.thelancet.com/diabetes-endocrinology Published online August 10, 2015 http://dx.doi.org/10.1016/S2213-8587(15)00261-2
one of the ligands for C-C chemokine receptor type 2
(CCR2), has been implicated, not only in insulin
resistance,9,10 but also in progressive renal injury and
has been suggested to be a potential marker of renal
disease.11 MCP-1 promotes monocyte and macrophage
migration and activation.12 CCX140-B is a small-
molecule CCR2 antagonist that inhibits CCR2 and
blocks MCP-1-dependent monocyte activation and
chemotaxis. Data from preclinical studies suggested
that oral CCX140-B improved glycaemia and
albuminuria in a mouse model of diabetes.13 Our aim in
this study was to test the e cacy and safety of two doses
of CCX140-B on albuminuria in patients with type 2
diabetes and proteinuria.
Methods
Study design and participants
We did this randomised, double-blind, placebo-controlled
trial at 78 research centres in Belgium, Czech Republic,
Germany, Hungary, Poland, and the UK. The initial study
included a 12-week treatment period because toxicology
results were not available at the time to support longer
term dosing. After long-term toxicology had been
successfully completed, we amended the protocol to
extend dosing from 12 weeks to 52 weeks with a 4-week
follow-up visit after stopping treatment. This protocol
amendment specifi ed that for patients already enrolled,
only those who completed the initial 12-week study
visit within the previous 16 weeks were eligible for
continuation to 52 weeks. Therefore, 95 of 332 patients
were not eligible to continue to 52 weeks because they
had been o study treatment for more than 16 weeks at
the time the protocol amendment was approved.
Eligible patients were aged 18–75 years inclusive and
had type 2 diabetes with proteinuria (fi rst morning
urine albumin to creatinine ratio [UACR] 100–3000 mg/g),
a glomerular fi ltration rate based on the modifi cation of
diet in renal disease equation (estimated glomerular
ltration rate [eGFR]) of greater than or equal to
25 mL/min per 1·73 m², a baseline HbA1c between
6–10% (4·2–8·6 mmol/mol), and a fasting plasma
glucose of less than 270 mg/dL (15 mmol/L). Patients
were on stable diabetes treatment and ACE inhibitors
or ARBs at a recommended therapeutic dose for at least
8 weeks.
Patients were excluded if they had type 1 diabetes or
known non-diabetic renal disease; a BMI higher than
45·4 kg/m²; cardiac failure (class III or IV), history of
unstable angina, symptomatic coronary artery disease,
myocardial infarction or stroke within 12 weeks before
screening; haemoglobin less than 10 g/dL (6·18 mmol/L);
or evidence of hepatic disease (aspartate amino-
transferase, alanine transaminase, or bilirubin >two
times the upper limit of normal). Patients were also
excluded if they had poorly controlled blood pressure
(systolic blood pressure >155 mm Hg or diastolic blood
pressure >95 mm Hg). Additional exclusion criteria
were use of bardoxolone, atrasentan, or other endothelin
antagonist within 8 weeks before screening; chronic
(>7 days continuously) non-steroidal anti-infl ammatory
drug treatment within 2 weeks before screening, or any
infection requiring antibiotic treatment within 4 weeks
before screening. The appendix contains the full list of
inclusion and exclusion criteria.
The trial was done in accordance with the Declaration
of Helsinki and Good Clinical Practice guidelines. Ethics
committees and institutional review boards approved the
research protocol. All patients gave written informed
consent before starting the trial.
Randomisation and masking
After the lead-in period, patients were stratifi ed based
on baseline UACR (100–300 mg/g, 301–800 mg/g, and
801–3000 mg/g) and baseline eGFR (25–59 mL/min per
1·73 m² and ≥60 mL/min per 1·73 m²), and then randomly
assigned (1:1:1) to receive oral placebo, CCX140-B 5 mg, or
CCX140-B 10 mg once a day for 52 weeks according to a
non-centre-specifi c randomi sation scheme. Stratifi cation
Research in context
Evidence before this study
We searched PubMed on May 4, 2015, and again on July 24,
2015, with the following search terms: “CCR2” AND “clinical
trial”, “CCR2” AND “diabetes”, and “CCR2” AND “diabetic
nephropathy”. This did not reveal any previous clinical trials
with any CCR2 drug in diabetic nephropathy. Before this clinical
trial, no other studies had been published for drugs targeting
the chemokine receptor CCR2 in patients with diabetic
nephropathy. One phase 2 study has been published with
CCX140-B in patients with type 2 diabetes. Patients in that
study did not have renal disease.
Added value of this study
To the best of our knowledge, the results from our trial provide
the fi rst evidence that CCR2 inhibition lowers albuminuria in
patients with type 2 diabetes on standard-of-care treatment,
with no major side-eff ects. These eff ects are achieved through
interference with a novel pathway compared with existing
treatments.
Implications of all the available evidence
Albuminuria lowering is surrogate evidence for renal
protection. The results of this trial showing that CCX140-B
lowers albuminuria should be translated into hard evidence in
follow-up studies that test whether CCX140-B also limits
progression to end-stage renal disease. If this is the case,
CCX140-B might have the potential to alter and attenuate the
relentless decline of renal function in patients with type 2
diabetes and proteinuria and thereby the need for expensive
renal replacement treatment.
Articles
www.thelancet.com/diabetes-endocrinology Published online August 10, 2015 http://dx.doi.org/10.1016/S2213-8587(15)00261-2
3
and randomisation was undertaken centrally via an
interactive web response system. The system used a
minimisation algorithm14 to maintain balance among the
three treatment groups with respect to the three baseline
UACR ranges, the two baseline eGFR ranges, and study
centre. Patients and all study personnel (except the safety
monitoring committee) were masked to treatment
allocation. CCX140-B is the sodium salt of CCX140. All
study drugs were in identical capsules, and supplied in
identical bottles labelled appropriately so as to maintain
the allocation masking.
Procedures
Patients who were randomised collected fi rst morning
void urine on 3 separate days at baseline and week 52,
and single measurements at weeks 2, 4, 8, 12, 16, 20, 28,
36, 44, and 56 for assessment of UACR. Urine albumin was
measured by a nephelometric assay and creatinine was
measured by a kinetic colorimetric assay in a central
laboratory (Medpace Reference Laboratory, Leuven,
Belgium). Serum creatinine, phosphorus, blood urea
nitrogen, and urinary MCP-1 to creatinine ratio were
measured at baseline and 2, 4, 8, 12, 16, 20, 28, 36, 44, 52,
and 56 weeks. Serum creatinine was used to calculate
eGFR with the modifi cation of diet in renal disease
equation:
eGFR=175 × (serum creatinine, mg/dL) ¹·¹ × (age, years)
·²³ × (0·742 if female) × (1·212 if black).
We measured HbA1c, fasting plasma glucose, and
homoeostatic model assessment of insulin resistance
(HOMA-IR) at baseline and 4, 8, 12, 20, 28, 36, 44, 52, and
56 weeks. HOMA-IR was calculated with the following
equation: fasting plasma glucose (mg/dL) × fasting
insulin (U/mL)/405. Plasma MCP-1 concentrations were
measured at baseline and weeks 12, 28, 52, and 56.
CCX140 plasma concentrations were measured at each
study visit using high-performance liquid chromatography
with tandem mass spectrometric detection (lower limit of
detection 1 ng/mL).
Safety was monitored at baseline and 2, 4, 8, 12, 20, 28,
36, 44, 52, and 56 weeks by assessing adverse events and
laboratory data. Reported adverse events were recorded
during the trial and analysed with a standard coding
dictionary (MedDRA, version 12.0) to classify adverse
event terms. Serious adverse events were defi ned as any
adverse event that resulted in death, was immediately life
threatening, required hospital admission, resulted in
persistent or substantial disability or incapacity, was a
birth defect, or was an important event that might heavily
jeopardise the patient or might have required intervention
to prevent any of the above.
We assessed patient compliance with taking study drug
based on returned capsule counts at each study visit as
well as plasma CCX140 concentrations measured over
the course of the trial.
Outcomes
The primary e cacy endpoint for the study was change
from baseline in UACR at 52 weeks. Because the treatment
period in the original protocol was 12 weeks, change
from baseline in UACR over 12 weeks was also analysed.
Secondary endpoints included change from baseline in
eGFR and HbA1c at 52 weeks. Other endpoints included
change from baseline in blood urea nitrogen, serum
phosphorus, fasting plasma glucose, fasting plasma
insulin, HOMA-IR, urinary MCP-1 to creatinine ratio
and plasma MCP-1 at week 52, and the plasma
concentration of CCX140 over the course of the study.
Statistical analysis
The planned sample size was at least 135 patients, 45 in
each group. However, the protocol made provision for an
enrolment target of 270 patients, 90 in each group, with
the anticipation that the trial dosing period might be
extended with potential loss of patients while the protocol
amendment was processed. Assuming a standard
deviation of 0·85, a sample size of 45 per group was
estimated to provide 80% power to detect a mean
between-group di erence in UACR change of 0·51,
corresponding to a 40% between-group di erence. A
sample size of 90 per group was estimated to provide
80% power to detect a mean between-group di erence in
UACR change of 0·36, corresponding to a 30% between-
group di erence.
We did e cacy analyses on the modifi ed intention-to-
treat population, which consisted of all patients with
uninterrupted dosing between the 12 week and 16 week
study visits. We excluded from the modifi ed intention-
to-treat population patients who stopped dosing at
week 12, either permanently, under the original protocol,
or temporarily, due to delay in protocol amendment
approval. The statistical analysis plan was revised
during the study to refl ect the dosing period extension
and change in primary endpoint (from week 12 to
week 52). We did safety analyses on all randomly
assigned patients who received at least one dose of study
drug. Analyses were based on comparisons between
placebo and CCX140-B groups as assigned by
randomisation, irrespective of whether patients followed
through the protocol or were fully compliant with the
protocol procedures.
The primary analysis was a mixed e ects model,
repeated measures (MMRM) analysis of change from
baseline to each post-baseline measurement of log UACR
using SAS (version 9.1). The model included treatment,
visit, and treatment-by-visit interaction as factors, and
baseline log UACR, eGFR, HbA1c, and mean arterial
pressure as covariates. UACR was log-transformed before
entering the data in the MMRM analysis to alleviate the
skewness of the data.
Visits were included as repeated measure units
from the same patients. To allow generality for the
covariance structure for the repeated measures, the
Articles
4
www.thelancet.com/diabetes-endocrinology Published online August 10, 2015 http://dx.doi.org/10.1016/S2213-8587(15)00261-2
variance-covariance matrix was assumed to be
unstructured—ie, purely data dependent. In this MMRM
model, all patients and all datapoints were included. No
patients were excluded because of missing data and no
imputation was done for missing data. We estimated the
treatment group di erence at week 52 with the simple
contrast and the overall between-group di erence over
the course of the study with the main e ects (contrast)
with the missing at random assumption. We compared
the contrast between each drug dose group and the
placebo group at week 52 with a prespecifi ed one-sided
signifi cance level of 0·05; treatment di erences at other
timepoints were also analysed.
The between-group geometric mean change (%) was
derived by 100*(exp [least squares mean change]–1), and
the same transformation was applied to the 95% confi dence
limits to obtain an approximate upper 95% confi dence
limit for the geometric mean change (%). The rationale for
one-sided testing for UACR was that in this phase 2 clinical
trial, the main goal was to test whether CCX140-B treatment
can reduce albuminuria. This was not a pivotal clinical trial,
in which a two-sided test would be more appropriate. We
used similar statistical models to assess treatment group
di erences in other e cacy and safety variables, such as
eGFR, HbA1c, and urinary MCP-1 to creatinine ratio. We
calculated two-sided 95% confi dence intervals for eGFR.
Figure 1: Trial profi le
UACR=urinary albumin to creatinine ratio. eGFR=estimated glomerular fi ltration rate. FPG=fasting plasma glucose. *Patient withdrawn because of discovery of
leukaemia medical history in the past (protocol deviation). †Patient withdrawn because of myocardial infarction and depression leading to diffi culty for patient to
comply with the protocol. ‡Patient withdrawn because of diagnosis of rectal cancer and diffi culty for patient to comply with the protocol.
883 patients screened
332 patients randomly assigned to treatment
111 assigned to placebo
(safety population)
110 assigned to 5 mg CCX140-B
(safety population)
111 assigned to 10 mg CCX140-B
(safety population)
10 discontinued
4 withdrew consent
5 adverse events (confusion,
balance disorder, heart valve
incompetence, prostate
cancer, myocardial infarction)
1 investigator decision*
6 discontinued
2 withdrew consent
4 adverse events (staphylococcal
bacteraemia, depression,
constipation, rash)
5 discontinued
3 withdrew consent
2 adverse events (anxiety,
diarrhoea)
106 completed 12 weeks 104 completed 12 weeks 101 completed 12 weeks
31 not eligible due to off study
drug for >16 weeks
3 withdrew consent
32 not eligible due to off study drug
for >16 weeks
4 withdrew consent
32 not eligible due to off study drug
for >16 weeks
64 uninterrupted dosing 63 uninterrupted dosing 65 uninterrupted dosing
59 completed 56 weeks 61 completed 56 weeks 62 completed 56 weeks
3 discontinued
2 withdrew consent
1 adverse event (atrial
fibrillation)
2 discontinued
1 relocation
1 investigator decision‡
5 discontinued
3 withdrew consent
1 adverse event (pneumonia)
1 investigator decision†
70 continued after 12 weeks 69 continued after 12 weeks 70 continued after 12 weeks
5 interrupted dosing6 interrupted dosing 6 interrupted dosing
551 ineligible
340 due to UAC R too low
39 due to HbA1c too low
26 due to UACR too high
24 due to eGFR too low
22 due to FPG too high
19 due to HbA1c too high
15 due to consent withdrawn
66 due to other inclusion or exclusion criteria
Articles
www.thelancet.com/diabetes-endocrinology Published online August 10, 2015 http://dx.doi.org/10.1016/S2213-8587(15)00261-2
5
A sensitivity analysis of change from baseline in log
UACR was done by an ANCOVA model with treatment as
a factor and baseline eGFR, HbA1c, and mean arterial
pressure as covariates, and last observation carried
forward imputation for missing data. Concomitant
drugs including RAS inhibitors, diuretics, β blockers,
calcium channel blockers, glucose-lowering drugs, and
lipid-modifying drugs were summarised by treatment
group.
The study was registered with ClinicalTrials.gov,
number NCT01447147.
Role of the funding source
The study was overseen by an advisory committee,
including members from the funder. The advisory
committee oversaw the design of the study, the conduct
of the trial, and the management and analysis of all data.
The funder was involved in the design of the study, in the
collection and analysis of the data, and in writing the
report. All authors had access to study results, and the
lead author takes responsibility for the accuracy and
completeness of the data reported. The lead author and
the advisory committee had the fi nal decision to submit
the publication.
Results
The study ran from Dec 7, 2011 (fi rst patient enrolled)
until Aug 4, 2014. We screened 883 patients of whom we
enrolled 332. 111 patients were assigned to placebo,
110 to CCX140-B 5 mg, and 111 to CCX140-B 10 mg.
209 patients continued after the 12-week period of whom
192 had uninterrupted treatment after week 12 (modifi ed
intention-to-treat population). 182 of the 192 patients
completed 56 weeks (fi gure 1).
Baseline demographics, clinical and biochemical
characteristics, and concomitant drugs were similar
between the three groups (table 1 and appendix). Apparent
slight baseline imbalances in average baseline plasma
MCP-1 concentrations are explained by one outlier value
each in the 5 mg and 10 mg CCX140-B group.
Repeated measures analysis showed a signifi cant least
squares mean change from baseline in albuminuria for
CCX140-B 5 mg (fi gure 2): –18% (95% CI –26% to
–8%; p=0·0004) compared with placebo –2% (95% CI
–11% to 9%; p=0·72) during 52 weeks; the di erence
between 5 mg CCX140-B and placebo was –16%
(one-sided 95% upper confi dence limit –5%; p=0·01).
The 10 mg dose showed a –11% (95% CI –20% to –1%;
p=0·02) least squares mean change from baseline
during 52 weeks; –10% (upper 95% confi dence limit 2%)
compared with placebo (p=0·08). Figure 2 also shows
the albuminuria changes in time. Geometric mean UACR
of 363 mg/g (95% CI 287–460) at baseline in the 5 mg
group was reduced to 276 mg/g (199–383) (–24% change;
p=0·0007) after 12 weeks of treatment, and remained
stable, ending at 296 mg/g (217–404) (–20% change;
p=0·03) at 52 weeks. The 10 mg dose of CCX140-B
showed a similar profi le of response in the fi rst 12 weeks,
–20% (95% CI –32 to –7) reduction compared with
baseline (p=0·005), but the e ect dissipated during the
subsequent weeks. For patients receiving placebo,
Placebo
(n=64)
5 mg CCX140-B
(n=63)
10 mg CCX140-B
(n=65)
Demographic characteristics
Age (years) 62·4 (7·6) 62·5 (8·0) 62·3 (7·9)
Women 15 (23%) 16 (25%) 14 (22%)
Ethnic origin
White 61 (95%) 62 (98%) 64 (98%)
Black 0 0 1 (2%)
Asian 3 (5%) 0 0
Pacifi c Islander 0 1 (2%) 0
Clinical characteristics
Weight (kg) 94·8 (18·6) 96·7 (13·7) 99·4 (17·4)
Known duration of diabetes (years) 14·9 (7·2) 15·0 (8·1) 16·3 (9·0)
Systolic blood pressure (mm Hg) 136·4 (15·2) 135·6 (14·5) 138·7 (14·2)
Diastolic blood pressure (mm Hg) 79·1 (9·2) 76·0 (9·0) 79·0 (8·4)
Serum albumin (g/L) 44·4 (3·2) 45·3 (3·7) 42·5 (8·0)
Serum creatinine (μmol/L)* 111·4 (42·5) 112·3 (42·0) 117·6 (47·3)
eGFR (mL/min per 1·73 m²) 64·2 (26·1) 62·4 (24·2) 61·1 (25·1)
Haemoglobin (g/L) 136·3 (13·4) 139·4 (16·2) 136·2 (15·9)
HbA1c (%) 7·7 (1·0) 7·5 (0·9) 7·8 (1·1)
Total cholesterol (mmol/L) 4·77 (1·05) 4·51 (1·12) 4·36 (1·00)
Serum potassium (mmol/L) 4·57 (0·48) 4·71 (0·50) 4·62 (0·49)
UACR (mg/g)† 440 (351–550) 363 (287–460) 438 (345–557)
Fasting plasma glucose (mmol/L) 8·79 (2·35) 9·46 (2·20) 9·46 (2·65)
Fasting plasma insulin (pmol/L) 210·4 (401·3) 150·7 (139·9) 181·3 (209·7)
HOMA-IR 13·0 (31·7) 9·5 (10·2) 11·9 (18·5)
Blood urea nitrogen (mmol/L) 8·50 (3·38) 9·25 (4·06) 9·03 (3·56)
Plasma MCP-1 (pg/mL) 218·3 (69·3) 283·0 (532·8) 229·3 (81·6)
Urine MCP-1 to creatinine ratio (pg/mg) 216·5 (310·5) 163·7 (120·4) 210·7 (165·9)
Drug treatments‡
Antihypertensives
ACE inhibitors 41 (64%) 45 (71%) 36 (55%)
Angiotensin receptor blockers 23 (36%) 18 (29%) 25 (39%)
ACE inhibitors plus angiotensin receptor
blockers
0 0 3 (5%)
β-blocking agents 29 (45%) 30 (48%) 29 (45%)
Dihydropyridine derivatives (calcium channel
blockers)
34 (53%) 34 (54%) 31 (48%)
Thiazides 10 (16%) 10 (16%) 11 (17%)
Drugs used in diabetes
Insulin and insulin analogues 33 (52%) 23 (37%) 29 (45%)
Biguanides (metformin) 36 (56%) 40 (64%) 33 (51%)
Sulphonamides, urea derivatives 18 (28%) 21 (33%) 15 (23%)
HMG CoA reductase inhibitors 40 (63%) 43 (68%) 40 (62%)
Data are mean (SD) or n (%) unless stated otherwise. The appendix shows characteristics of all randomised patients.
ACE=angiotensin-converting enzyme. eGFR=estimated glomerular fi ltration rate. UACR=urine albumin to urine
creatinine ratio. MCP-1=monocyte chemoattractant protein-1. HOMA-IR=homoeostatic model assessment of insulin
resistance. HMG CoA=3-hydroxy-3-methyl-glutaryl-CoA. *To convert to mg/dL, divide by 88·4. †Geometric mean and
95% CI. ‡Baseline drug treatments were summarised by selected Anatomical Therapeutic Chemical (ATC) classifi cation
system codes using WHO dictionary (version 9.1).
Table 1: Baseline characteristics of patients in the modifi ed intention-to-treat population
Articles
6
www.thelancet.com/diabetes-endocrinology Published online August 10, 2015 http://dx.doi.org/10.1016/S2213-8587(15)00261-2
baseline UACR showed no signifi cant change over time.
4 weeks after drug discontinuation, geometric mean
UACR remained at similar levels as the in-trial results for
all groups (fi gure 2). Results from a sensitivity analysis
using an ANCOVA model were consistent with the
MMRM analysis (appendix). The UACR response to
CCX140-B showed no interactions with the di erent
baseline parameters except for HbA1c, although the latter
with no consistent pattern (appendix). Because many
patients who completed the original 12-week protocol
were ineligible to participate in the 52-week protocol, we
assessed the results for all randomly assigned patients to
assess whether the results were consistent. The
albuminuria change at week 12 was –18% (upper 95%
confi dence limit –7%; p=0·006) for the 5 mg CCX140-B
and –4% (upper 95% confi dence limit 9%; p=0·29) for
the 10 mg group compared with placebo. The baseline
characteristics of all randomly assigned patients versus
the 52-week population showed no meaningful
di erences (appendix), suggesting that the 52-week
population was representative of the randomised
population.
Changes in eGFR, HbA1c, fasting plasma insulin,
HOMA-IR, serum phosphorus, blood urea nitrogen,
blood pressure, and urinary MCP-1 in participants who
received CCX140-B were not signifi cant compared with
those who received placebo (table 2 and appendix). There
was a greater change in fasting plasma glucose in
the 5 mg CCX140-B group compared with placebo
(–1·12 mmol/L, upper 95% confi dence limit –0·24;
Figure 2: Primary endpoint
Urinary albumin to creatinine ratio change in percent geo metric mean from baseline to 56 weeks for patients receiving placebo, CCX140-B 5 mg, or CCX140-B 10 mg.
Least-squares means change (±95% CIs) in urinary albumin to creatinine ratio (integral response) are plotted on the right hand side of the fi gure and represent the
primary endpoint. *p=0·01 for CCX140-B compared with placebo.
Number at risk
Placebo
5 mg CCX140-B
10 mg CCX140-B
0 4 8 12 16 20 24 28 32 36 40 44 48 52
*
Recovery
Least-squares
mean
56
64
63
65
64
63
63
64
63
65
64
62
62
62
60
59
62
58
57
62
59
57
59
59
56
59
57
57
58
58
56
58
58
52
58
58
57
Time (weeks)
–30
–25
–20
–15
–10
–5
0
5
10
15
Albuminuria change from baseline (%)
Placebo
5 mg CCX140-B
10 mg CCX140-B
Placebo 5 mg CCX140-B 10 mg CCX140-B
n Least-squares mean
(95% CI)
n Least-squares mean
(95% CI)
p value vs
placebo
n Least-squares mean
(95% CI)
p value vs
placebo
eGFR (mL/min/1·73 m²) 57 –2·6 (–4·6 to –0·6) 58 –2·4 (–4·4 to –0·4) 0·88 52 –3·8 (–5·9 to –1·8) 0·39
HbA1c (%) 58 0·12 (–0·08 to 0·33) 57 0·16 (–0·05 to 0·36) 0·58 52 –0·08 (–0·29 to 0·13) 0·08
Fasting plasma glucose (mmol/L) 58 0·38 (–0·36 to 1·11) 56 –0·74 (–1·49 to 0·01) 0·01 52 –0·37 (–1·13 to 0·39) 0·08
Fasting plasma insulin (pmol/L) 58 –48·9 (–106·6 to 8·8) 56 –14·5 (–73·0 to 44·1) 0·40 52 –0·1 (–59·7 to 59·5) 0·24
HOMA-IR 58 –2·14 (–6·59 to 2·31) 56 –1·37 (–5·88 to 3·14) 0·59 52 –1·20 (–5·77 to 3·38) 0·61
Phosphorus (mmol/L) 57 0·06 (0·03 to 0·10) 58 0·03 (–0·01 to 0·07) 0·20 52 0·03 (–0·01 to 0·07) 0·26
Blood urea nitrogen (mmol/L) 57 0·34 (–0·27 to 0·96) 58 0·49 (–0·13 to 1·11) 0·74 52 –0·07 (–0·71 to 0·57) 0·35
Plasma MCP-1 (pg/mL) 58 42·3 (4·4 to 80·3) 58 69·7 (32·1 to 107·3) 0·31 52 111·0 (69·6 to 152·5) 0·01
Urine MCP-1 to creatinine ratio (%) 57 26 (3 to 55) 57 46 (19 to 79) 0·84 47 29 (3 to 61) 0·55
Plasma CCX140 at week 52 (ng/mL), mean (SD) .. .. 57 1265 (379) NA 55 2299 (1057) NA
Data are least-squares mean (95% CI) unless otherwise noted. HOMA-IR=homoeostasis model assessment of insulin resistance; MCP-1=monocyte chemoattractant protein-1; NA=not applicable. Plasma CCX140
concentrations were not measured in the placebo group. p values are from the mixed model of repeated measures analysis including treatment, visit, treatment-by-visit interaction as factors, and baseline log
UACR, eGFR, HbA1c, and mean arterial pressure as covariates.
Table 2: Summary of change from baseline to week 52 by treatment group for secondary and other study endpoints in the modifi ed intention-to-treat population
Articles
www.thelancet.com/diabetes-endocrinology Published online August 10, 2015 http://dx.doi.org/10.1016/S2213-8587(15)00261-2
7
p=0·01). The average plasma concentration of CCX140
was roughly twice as high in the 10 mg CCX140-B group
compared with the 5 mg CCX140-B group, and remained
stable over the course of the study (appendix). The
average plasma MCP-1 concentration rose signifi cantly
in the 10 mg CCX140-B group but not in the other two
groups (table 2 and fi gure 3A). The UACR response
(baseline to week 52) seemed to be attenuated in the
presence of higher rise in plasma levels of MCP-1 upon
treatment (fi gure 3B).
The study drug was generally well tolerated (table 3).
Adverse events were consistent with the age and
underlying medical conditions of the patient population.
We noted that serious adverse events occurred in a
similar proportion of participants in the placebo and
5 mg CCX140-B groups (13 [11·7%] in the placebo group
and 13 [11·8%] in the 5 mg CCX140-B group). The 10 mg
CCX140-B group had a higher incidence of serious
adverse events: 25 (22·5%). Two serious adverse events
in the placebo group, and one each in the 5 mg and
10 mg CCX140-B groups, were reported to possibly be
related to the study drug: complete atrioventricular block
and myocardial infarction; Staphylococcal bacteraemia
after tooth extraction (5 mg CCX140-B) and subcutaneous
abscess (10 mg CCX140-B). Two patients died during the
course of the study, neither deemed drug related by the
investigators (one due to stroke in the 5 mg CCX140-B
group and one due to myocardial infarction in the 10 mg
CCX140-B group). None of the patients had a renal event:
no confi rmed doubling of serum creatinine over the
course of the study, none underwent dialysis, and none
had an eGFR of less than 15 mL/min per 1·73 m².
Patient compliance with taking study drug was high:
108 (97%) of 111 in the placebo group, 108 (98%) of 110 in
the 5 mg CCX140-B group, and 108 (97%) of 111 patients
in the 10 mg CCX140-B group took more than 90% of
their prescribed study drug. Compliance was further
substantiated by the plasma CCX140 concentration
measurements over the course of the study. Six (3%) of
221 patients, three in each of the CCX140-B groups, had
evidence of non-compliance based on plasma CCX140
concentrations.
Discussion
In patients with type 2 diabetes with proteinuria who
were on a therapeutic dose of ACE inhibitor or ARB, a
dose of 5 mg CCX140-B lowered albuminuria by 18%
overall versus 2% with placebo during the course of the
study without major side-e ects. The magnitude of this
e ect is deemed clinically meaningful and could translate
into further renal benefi t with long-term treatment.
Recently the focus of renoprotective treatments has
switched from targeting haemodynamic pathways to
infl ammatory pathways. Further inhibition of the RAS
seems to cause increased side-e ects that might counteract
the benefi cial e ects of reducing classical risk factors like
glucose, blood pressure, and albuminuria. Targeting other
pathways might be a better option for enhanced renal
protection in advanced type 2 diabetes. The chemokine
receptor 2 pathway seems interesting in view of the fact
that this pathway has been associated with progressive
renal function loss,11 and that inhibition of CCR2 a ords
renal protection in experimental models of renal disease.13
This is the fi rst study describing the e ect of CCR2
inhibition on renal risk parameters such as albuminuria,
in patients with type 2 diabetes and nephropathy.
The exact mechanism of the renoprotective e ect
of CCR2 inhibition is unknown. Several possibilities
have been discussed11,13,15,16 including blocking renal
macrophage infi ltration, infl ammation, oxidative stress,
improving podocyte number and function, and
interaction with the RAS. Our trial was designed to
Figure 3: MCP-1 response and the relation with UACR
(A) Least squares mean (95% CI) change in plasma MCP-1 levels in the placebo, 5 mg CCX140-B group, and 10 mg CCX140-B group at week 52. (B) Geometric mean
(95% CI) albuminuria change versus the geometric mean change (95% CI) in MCP-1 change (in tertiles) from baseline to week 52 for the 5 mg CCX140-B group and
10 mg CCX140-B group combined (p for trend [continuous]=0·03). Median plasma concentrations of MCP-1 tertiles at week 52 are 257 pg/mL in the lower tertile,
279 pg/mL in the middle tertile, and 353 pg/mL in the upper tertile. UACR=urinary albumin to creatinine ratio. *p=0·01 for CCX140-B compared with placebo.
Placebo
5 mg CCX140-B 10 mg CCX140-B
0
20
40
60
80
100
120
140
160
180
Change in plasma MCP-1 concentration (pg/mL)
A
–20 0 20 40 60 80 100 120 140
–60
–40
–20
0
20
*
40
Albuminuria change (%)
MCP-1 change (%)
B
Articles
8
www.thelancet.com/diabetes-endocrinology Published online August 10, 2015 http://dx.doi.org/10.1016/S2213-8587(15)00261-2
inform the design and study population for a potential
phase 3 trial. It was not designed to explore the
mechanism by which CCX140-B lowers albuminuria,
and thus does not further detail the mechanism of
CCR2 inhibition in renal protection. We selected
CCX140-B doses of 5 mg and 10 mg once a day because
both result in plasma concentrations that block
CCR2-mediated cell migration at about 90% or greater,13
and both doses were found to be safe and well tolerated
in an earlier study in patients with type 2 diabetes.17
Placebo
(n=111)
5 mg
CCX140-B
(n=110)
10 mg
CCX140-B
(n=111)
Patients with any adverse event 81 (73%) 71 (65%) 68 (61%)
Patients with any serious
adverse event
13 (12%) 13 (12%) 25 (23%)
Patients with adverse events
leading to discontinuation of
study drug
3 (3%) 6 (6%) 6 (5%)
Renal events
Doubling of serum creatinine 0 0 0
eGFR <15 mL/min per
1·73 m²
00 0
Dialysis or transplantation 0 0 0
Death 0 1 (<1%) 1 (<1%)
All patients with serious adverse events per system organ class
Infections and infestations 4 (4%) 4 (4%) 6 (5%)
Pneumonia 2 (2%) 1 (<1%) 1 (<1%)
Cellulitis 0 0 1 (<1%)
Gangrene 0 0 1 (<1%)
Lobar pneumonia 0 0 1 (<1%)
Localised infection 0 0 1 (<1%)
Lower respiratory infection 0 1 (<1%) 0
Respiratory tract infection 0 0 1 (<1%)
Septic shock 0 0 1 (<1%)
Staphylococcal
bacteraemia
0 1 (<1%) 0
Subcutaneous abscess 1 (<1%) 0 1 (<1%)
Urinary tract infection 0 1 (<1%) 0
Lung infection 1 (<1%) 0 0
Cardiac disorders 3 (3%) 2 (2%) 4 (4%)
Angina unstable 0 2 (2%) 1 (<1%)
Atrial fi brillation 0 0 1 (<1%)
Cardiac failure 1 (<1%) 0 1 (<1%)
Coronary artery stenosis 0 1 (<1%) 0
Myocardial infarction 1 (<1%) 0 1 (<1%)
Atrioventricular block
complete
1 (<1%) 0 0
Gastrointestinal disorders 2 (2%) 1 (<1%) 4 (4%)
Abdominal pain 0 0 1 (<1%)
Duodenal ulcer 0 0 1 (<1%)
Gastrointestinal
haemorrhage
0 0 1 (<1%)
Nausea 0 0 1 (<1%)
Refl ux oesophagitis 0 1 (<1%) 0
Constipation 1 (<1%) 0 0
Diarrhoea 1 (<1%) 0 0
Metabolism and nutrition
disorders
0 2 (2%) 2 (2%)
Hyperglycaemia 0 1 (<1%) 1 (<1%)
Diabetes mellitus 0 0 1 (<1%)
Hypoglycaemia 0 1 (<1%) 0
(Table 3 continues in next column)
Placebo
(n=111)
5 mg
CCX140-B
(n=110)
10 mg
CCX140-B
(n=111)
(Continued from previous column)
Neoplasms benign,
malignant and unspecifi ed
2 (2%) 1 (<1%) 3 (3%)
Renal cancer 0 0 2 (2%)
Prostate cancer 0 0 1 (<1%)
Rectal cancer stage IV 0 1 (<1%) 0
Bladder transitional cell
carcinoma
1 (<1%) 0 0
Meningioma 1 (<1%) 0 0
Nervous system disorders 1 (<1%) 2 (2%) 2 (2%)
Ischaemic stroke 0 1 (<1%) 1 (<1%)
Balance disorder 0 0 1 (<1%)
Cerebrovascular accident 0 1 (<1%) 0
Subarachnoidal
haemorrhage
0 0 1 (<1%)
Cerebral infarction 1 (<1%) 0 0
Vascular disorders 2 (2%) 1 (<1%) 2 (2%)
Deep vein thrombosis 0 0 1 (<1%)
Femoral arterial stenosis 0 0 1 (<1%)
Hypertension 0 1 (<1%) 0
Peripheral vascular disorder 1 (<1%) 0 0
Subclavian steal syndrome 1 (<1%) 0 0
Renal and urinary disorders 1 (<1%) 1 (<1%) 1 (<1%)
Renal failure acute 1 (<1%) 1 (<1%) 1 (<1%)
Respiratory, thoracic and
mediastinal disorders
1 (<1%) 1 (<1%) 1 (<1%)
Chronic obstructive
pulmonary disease
1 (<1%) 1 (<1%) 0
Pulmonary embolism 0 0 1 (<1%)
Skin and subcutaneous
tissue disorders
0 0 2 (2%)
Angioedema 0 0 1 (<1%)
Purpura senile 0 0 1 (<1%)
Eye disorders 0 0 1 (<1%)
Eye haemorrhage 0 0 1 (<1%)
Injury, poisoning, and
procedural complications
2 (2%) 0 1 (<1%)
Fall 0 0 1 (<1%)
Spinal compression
fracture
1 (<1%) 0 0
Tendon rupture 1 (<1%) 0 0
Musculoskeletal and
connective tissue disorders
1 (<1%) 0 0
Meniscal degeneration 1 (<1%) 0 0
Data are n (%). eGFR=estimated glomerular fi ltration rate.
Table 3: Adverse events in the safety population
Articles
www.thelancet.com/diabetes-endocrinology Published online August 10, 2015 http://dx.doi.org/10.1016/S2213-8587(15)00261-2
9
CCX140-B improves proteinuria and glycaemia in
mice13 and our study points to similar e ects in human
beings. Could the e ect on proteinuria and glycaemia be
related? In mice, the e ect on proteinuria was noted
earlier after treatment onset, and at lower doses of the
CCR2 inhibitor, than was the e ect on glycaemia.13 The
benefi cial e ect of CCR2 inhibition on glycaemic control
is thought to be related to a reduction in the macrophage
content in adipose tissue, leading to improved insulin
sensitivity.9,10 The e ect on proteinuria is probably caused
by other mechanisms, such as reduced infl ammation or
improved podocyte integrity.
An important fi nding was the apparent lack of a dose
response in our study. The 10 mg dose seemed to have
less albuminuria-lowering e ect than 5 mg CCX140-B.
The drug was adequately absorbed, as participants in the
10 mg CCX140-B group had twice the plasma drug levels
as the 5 mg dose. However, we also noted that
concentrations of an endogenous ligand for CCR2,
MCP-1 (also known as CCL2) rose with increasing dose
of the drug. Within each of the groups that received
CCX140-B, we noted that higher MCP-1 levels were
associated with less reduction in albuminuria, especially
in the 10 mg group.
We hypothesise that the increased concentrations of
MCP-1 compete with the drug. This is consistent with
reaching the plateau of the dose response with
10 CCX140-B mg at 12 weeks and attenuation of the
albuminuria-lowering e ect after week 12 in the 10 mg
CCX140-B group. Compliance as shown by the reported
taking of study drug was high and similar across groups.
Additionally, CCX140 plasma concentrations were
consistent across all study visits. The dropout rate in the
5 mg and 10 mg CCX140-B groups was similar. Therefore,
non-compliance with study drug or patient dropout were
unlikely to explain the attenuation of the drug’s e ect after
week 12 in the 10 mg group.
Our study design has limitations. In particular, the
study was amended from a 12-week study to a 52-week
study; not all patients enrolled in the 12-week study
could continue into the 52-week study because at the
crucial time of re-enrolment into the study extension,
the long-term toxicology data were not yet available.
This resulted in a loss of patients that had been exposed
to drug or placebo without interruption between
weeks 12 and 16. However, UACR results at week 12 for all
randomly assigned patients were consistent with the
results from patients with uninterrupted treatment
after week 12, suggesting that we did not selectively lose
patients.
What are the chances that 5 mg CCX140-B will be
renoprotective and delay hard renal endpoints? We noted
that changes in eGFR were not markedly di erent
between the placebo group and the 5 mg CCX140-B after
1 year. However, a di erence was not expected in view of
the the fact that the study was not statistically powered
for this endpoint. Moreover previous hard outcome trials
have shown renal protection in end stage renal disease
endpoints without marked changes in eGFR slopes
during 1 year.18 The fi nding that CCX140-B lowered
albuminuria by 16% compared with placebo, on top of
standard of care, could be an indicator of potential renal
protection. Findings of a recent meta-analysis showed
that all interventions that lower albuminuria by more
than 15% in the fi rst months of treatment are associated
with an improvement in hard renal outcomes compared
with standard of care.19 Whether this will hold true for
CCX140-B, which has a novel mechanism of action
compared with existing treatments, is unknown. The
current profi le of CCX140-B does not show any emerging
side-e ects such as hyperkalaemia or cardiovascular
events that could o set its benefi cial renoprotective
properties.
In conclusion, 5 mg CCX140-B once a day lowers
albuminuria in patients with type 2 diabetes and
proteinuria, on top of standard of care, and without any
marked side-e
ects. These results suggest that
CCX140-B is a promising candidate for further clinical
development to reduce the unmet need for treatments
for diabetic renal disease.
Contributors
PB and TJS designed the study with scientifi c advisers. PB and AP ran
the study with their teams. EH, CH, IG-B, HM, and VT enrolled patients
and critically reviewed the manuscript. DdZ, PB, and HJLH performed
the data analysis and statistical analysis. All authors interpreted the data.
DdZ wrote the draft of the report and all authors contributed to its
revisions. DdZ takes responsibility for the full report. The appendix lists
all investigators who enrolled patients in the trial.
Declaration of interests
DdZ is a consultant for and receives honoraria (to employer) from
AbbVie, Astellas, ChemoCentryx, Eli Lilly, Janssen, Fresenius,
Merck Darmstadt. PB, AP, and TJS are employees and shareholders of
ChemoCentryx, the funder of this study. IG-B has received research
grants from Bayer HealthCare; consultant, advisory board or lecture fees,
and travel expenses from Genzyme, MSD, Novartis, Novo Nordisk,
Pfi zer, Ipsen, Bristol-Myers Squibb, AstraZeneca, Aegereon, Amgen,
Sanofi , Otsuka, Lilly, Chiesi, and ChemoCentryx. EH, CH, and HM have
no declarations regarding potential confl ict of interest. VT obtained
lecture fees from Roche, Medonet, Baxter, Fresenius Medical Care,
B. Braun, and Amgen, and consulting fees from AbbVie, ChemoCentryx,
and Boehringer Ingelheim. He has served as a member of the Advisory
Board of AbbVie, Amgen, Fresenius Medical Care, and Baxter. HJLH
received a research grant from AstraZeneca (payment to employer) and
is a consultant for and receives honoraria (to employer) from AbbVie,
Astellas, Janssen, Reata Pharmaceuticals, and ZS-Pharma.
Acknowledgments
This study was funded by ChemoCentryx. We thank all study
coordinators, investigators, and patients for their valuable contribution.
We thank Chao Wang (Pharma Data Associates), Sharon Barker
(Medpace), and Tobias Kröpelin (University Medical Centre
Groningen), who helped with statistical analysis; and Israel Charo for a
critical review of the report.
References
1 de Zeeuw D, Remuzzi G, Parving HH, et al. Proteinuria, a target
for renoprotection in patients with type 2 diabetic nephropathy:
lessons from RENAAL. Kidney Int 2004; 65: 2309–20.
2 Atkins RC, Briganti EM, Lewis JB, et al. Proteinuria reduction and
progression to renal failure in patients with type 2 diabetes mellitus
and overt nephropathy. Am J Kidney Dis 2005; 45: 281–87.
3 Cravedi P, Ruggenenti P, Remuzzi G. Proteinuria should be used as
a surrogate in CKD. Nat Rev Nephrol 2012; 8: 301–06.
Articles
10
www.thelancet.com/diabetes-endocrinology Published online August 10, 2015 http://dx.doi.org/10.1016/S2213-8587(15)00261-2
4 Ibsen H, Olsen MH, Wachtell K, Borch-Johnsen K, Lindholm LH,
Mogensen CE. Reduction in albuminuria translates to reduction in
cardiovascular events in hypertensive patients with left ventricular
hypertrophy and diabetes. J Nephrol 2008; 21: 566–69.
5 Schmieder RE, Mann JF, Schumacher H, et al, and the ONTARGET
Investigators. Changes in albuminuria predict mortality and
morbidity in patients with vascular disease. J Am Soc Nephrol 2011;
22: 1353–64.
6 Heerspink HJ, de Zeeuw D. The kidney in type 2 diabetes therapy.
Rev Diabet Stud 2011; 8: 392–402.
7 Parving HH, Brenner BM, McMurray JJ, et al, and the ALTITUDE
Investigators. Cardiorenal end points in a trial of aliskiren for type 2
diabetes. N Engl J Med 2012; 367: 2204–13.
8 Fried LF, Emanuele N, Zhang JH, et al, and the VA NEPHRON-D
Investigators. Combined angiotensin inhibition for the treatment of
diabetic nephropathy. N Engl J Med 2013; 369: 1892–903.
9 Sartipy P, Loskuto DJ. Monocyte chemoattractant protein 1 in obesity
and insulin resistance. Proc Natl Acad Sci USA 2003; 100: 7265–70.
10 Kanda H, Tateya S, Tamori Y, et al. MCP-1 contributes to
macrophage infi ltration into adipose tissue, insulin resistance, and
hepatic steatosis in obesity. J Clin Invest 2006; 116: 1494–505.
11 Tesch GH. MCP-1/CCL2: a new diagnostic marker and therapeutic
target for progressive renal injury in diabetic nephropathy.
Am J Physiol Renal Physiol 2008; 294: F697–701.
12 Charo IF, Myers SJ, Herman A, et al. Molecular cloning and
functional expression of two monocyte chemoattractant protein 1
receptors reveals alternative splicing of the carboxyl-terminal tails.
Proc Natl Acad Sci USA 1994; 91: 2752–56.
13 Sullivan T, Miao Z, Dairaghi DJ, et al. CCR2 antagonist CCX140-B
provides renal and glycemic benefi ts in diabetic transgenic human
CCR2 knockin mice. Am J Physiol Renal Physiol 2013; 305: F1288–97.
14 Pocock SJ, Simon R. Sequential treatment assignment with
balancing for prognostic factors in the controlled clinical trial.
Biometrics 1975; 31: 103–15.
15 Ayoub MA, Zhang Y, Kelly RS, et al. Functional Interaction between
Angiotensin II Receptor Type 1 and Chemokine (C-C Motif)
Receptor 2 with Implications for Chronic Kidney Disease. PLoS One
2015; 10: e0119803.
16 Seok SJ, Lee ES, Kim GT, et al. Blockade of CCL2/CCR2 signalling
ameliorates diabetic nephropathy in db/db mice.
Nephrol Dial Transplant 2013; 28: 1700–10.
17 Hanefeld M, Schell E, Gouni-Berthold I, et al. Orally-administered
chemokine receptor CCR2 antagonist CCX140-B in type 2 diabetes:
a pilot double-blind, randomized clinical trial. J Diabetes Metab
2012; 3: 225. DOI:10.4172/2155-6156.1000225.
18 Holtkamp FA, de Zeeuw D, Thomas MC, et al. An acute fall in
estimated glomerular fi ltration rate during treatment with losartan
predicts a slower decrease in long-term renal function. Kidney Int
2011; 80: 282–87.
19 Lambers Heerspink HJ, Kröpelin TF, Hoekman J, et al, and the
Reducing Albuminuria as Surrogate Endpoint (REASSURE)
Consortium. Drug-induced reduction in albuminuria is associated
with subsequent renoprotection: a meta-analysis. J Am Soc Nephrol
2014; published Nov 24. DOI:10.1681/ASN.2014070688.
... Members of class A (rhodopsin) G protein-coupled receptor (GPCR) superfamily, chemokine receptors have been considered promising drug targets due to involvement in cancer pathogenesis, inflammatory disorders, and other immune system dysregulations [2][3][4]. Notably, CC chemokine receptor 2 (CCR2), along with its cognate chemokine CCL2, displayed potential as a target in cardiovascular [5] disease, diabetes [6][7][8][9], liver fibrosis [10,11], neuropathic pain [12], rheumatoid arthritis [13], neuroinflammation [14,15], and carcinogenesis [16][17][18]. More recently, there has been a great interest in inhibition of CCR2-mediated immunosuppression in the context of immuno-oncology [19][20][21][22][23][24][25]. ...
... However, the only approval to date has been maraviroc, a CCR5 antagonist, for the treatment of HIV infection [33][34][35][36]. Several CCR2 antagonists have shown mixed results in early stage clinical trials [6,9,[37][38][39][40][41][42][43][44][45][46]. ...
... Here, using molecular docking and virtual compound library screening, we systematically identify the determinants of binding affinity and receptor and species selectivity for various chemotypes of orthosteric and allosteric antagonists of CCR2. The generated 3D models of receptor complexes with selective and dual-affinity antagonists, including the former clinical candidates cenicriviroc [45], and CCX-140 [9] and the current clinical candidates BMS-813160 [69,70] and PF-4136309 [42], reveal hitherto unappreciated mechanisms of antagonist affinity and selectivity. Findings are further corroborated by insights from compound structure-activity relationship (SAR) series and rationally guided gain-of-function receptor mutagenesis. ...
Preprint
Full-text available
By driving monocyte chemotaxis, the chemokine receptor CCR2 shapes inflammatory responses and the formation of tumor microenvironments. This makes it a promising target in inflammation and immuno-oncology. Unfortunately, despite extensive efforts, no CCR2-targeting therapeutics have yet reached the clinic. Cited reasons include the redundancy of the chemokine system, suboptimal properties of compound candidates, and poor agreement of clinical responses with preclinical murine model studies. Structure-based drug design approaches can rationalize and greatly accelerate CCR2 compound discovery and optimization. The prerequisites for such efforts include a good atomic-level understanding of the molecular determinants of action of existing antagonists. In this study, using molecular docking and artificial-intelligence-powered compound library screening, we uncover the structural principles of small molecule antagonism and selectivity towards CCR2 and its sister receptor CCR5. We show that CCR2 orthosteric inhibitors universally occupy an inactive-state-specific tunnel between receptor helices 1 and 7; we also discover an unexpected role for an extra-helical groove accessible through this tunnel, suggesting its potential as a new targetable interface for CCR2 and CCR5 modulation. We implicate a single CCR2 residue, S101 2.63 , as a determinant of CCR2/CCR5 and human/mouse antagonist selectivity, and corroborate its role through experimental gain-of-function mutagenesis. We systematically identify the binding determinants for various chemotypes of allosteric antagonists. We establish a critical role of induced fit in antagonist recognition, reveal strong chemotype selectivity of existing structures, and demonstrate the high predictive potential of a new deep-learning-based compound scoring function. Finally, we expand the available CCR2 structural landscape with computationally generated chemotype-specific models well-suited for structure-based antagonist design.
... In the translational process, clinical trials can provide valuable insight into the safety of utilizing chemokine therapeutics in other diseases. For instance, studies have investigated the prevention of diabetic nephropathy progression through the blocking of monocyte recruitment with small molecules, such as a specific CCR2 or dual CCR2/CCR5 antagonist (160,161). Applying these antagonists to human subjects for either twelve or fifty-two weeks appears to be safe, with a notable decrease in circulating monocytes and a concomitant increase in CCL2 blood concentrations. ...
Article
Full-text available
Blood clotting is a finely regulated process that is essential for hemostasis. However, when dysregulated or spontaneous, it promotes thrombotic disorders. The fact that these are triggered, accompanied and amplified by inflammation is reflected in the term thromboinflammation that includes chemokines. The role of chemokines in thrombosis is therefore illuminated from a cellular perspective, where endothelial cells, platelets, red blood cells, and leukocytes may be both the source and target of chemokines. Chemokine-dependent prothrombotic processes may thereby occur independently of chemokine receptors or be mediated by chemokine receptors, although the binding and activation of classical G protein-coupled receptors and their signaling pathways differ from those of atypical chemokine receptors, which do not function via cell activation and recruitment. Regardless of binding to their receptors, chemokines can induce thrombosis by forming platelet-activating immune complexes with heparin or other polyanions that are pathognomonic for HIT and VITT. In addition, chemokines can bind to NETs and alter their structure. They also change the electrical charge of the cell surface of platelets and interact with coagulation factors, thereby modulating the balance of fibrinolysis and coagulation. Moreover, CXCL12 activates CXCR4 on platelets independently of classical migratory chemokine activity and causes aggregation and thrombosis via the PI3Kβ and Btk signaling pathways. In contrast, typical chemokine-chemokine receptor interactions are involved in the processes that contribute to the adhesiveness of the endothelium in the initial phase of venous thrombosis, where neutrophils and monocytes subsequently accumulate in massive numbers. Later, the reorganization and resolution of a thrombus require coordinated cell migration and invasion of the thrombus, and, as such, indeed, chemokines recruit leukocytes to existing thrombi. Therefore, chemokines contribute in many independent ways to thrombosis.
Article
Full-text available
Macrophages are exceptionally diversified cell types and perform unique features and functions when exposed to different stimuli within the specific microenvironment of various kidney diseases. In instances of kidney tissue necrosis or infection, specific patterns associated with damage or pathogens prompt the development of pro-inflammatory macrophages (M1). These M1 macrophages contribute to exacerbating tissue damage, inflammation, and eventual fibrosis. Conversely, anti-inflammatory macrophages (M2) arise in the same circumstances, contributing to kidney repair and regeneration processes. Impaired tissue repair causes fibrosis, and hence macrophages play a protective and pathogenic role. In response to harmful stimuli within the body, inflammasomes, complex assemblies of multiple proteins, assume a pivotal function in innate immunity. The initiation of inflammasomes triggers the activation of caspase 1, which in turn facilitates the maturation of cytokines, inflammation, and cell death. Macrophages in the kidneys possess the complete elements of the NLRP3 inflammasome, including NLRP3, ASC, and pro-caspase-1. When the NLRP3 inflammasomes are activated, it triggers the activation of caspase-1, resulting in the release of mature proinflammatory cytokines (IL)-1β and IL-18 and cleavage of Gasdermin D (GSDMD). This activation process therefore then induces pyroptosis, leading to renal inflammation, cell death, and renal dysfunction. The NLRP3–ASC–caspase-1–IL-1β–IL-18 pathway has been identified as a factor in the development of the pathophysiology of numerous kidney diseases. In this review, we explore current progress in understanding macrophage behavior concerning inflammation, injury, and fibrosis in kidneys. Emphasizing the pivotal role of activated macrophages in both the advancement and recovery phases of renal diseases, the article delves into potential strategies to modify macrophage functionality and it also discusses emerging approaches to selectively target NLRP3 inflammasomes and their signaling components within the kidney, aiming to facilitate the healing process in kidney diseases.
Article
Full-text available
Diabetes mellitus (DM) often causes chronic kidney damage despite best medical practices. Diabetic kidney disease (DKD) arises from a complex interaction of factors within the kidney and the whole body. Targeting specific disease-causing agents using drugs has not been effective in treating DKD. However, stem cell therapies offer a promising alternative by addressing multiple disease pathways and promoting kidney regeneration. Mesenchymal stem cells (MSCs) offer great promise due to their superior accessibility ratio from adult tissues and remarkable modes of action, such as the production of paracrine anti-inflammatory and cytoprotective substances. This review critically evaluates the development of MSC treatment for DKD as it moves closer to clinical application. Results from animal models suggest that systemic MSC infusion may positively impact DKD progression. However, few registered and completed clinical trials exist, and whether the treatments are effective in humans is still being determined. Significant knowledge gaps and research opportunities exist, including establishing the ideal source, dose, and timing of MSC delivery, better understanding of in vivo mechanisms, and developing quantitative indicators to obtain a more significant therapeutic response. This paper reviews recent literature on using MSCs in preclinical and clinical trials in DKD. Potent biomarkers related to DKD are also highlighted, which may help better understand MSCs’ action in this disease progression. Key messages Mesenchymal stem cells have anti-inflammatory and paracrine effects in diabetic kidney disease. Mesenchymal stem cells alleviate in animal models having diabetic kidney disease. Mesenchymal stem cells possess promise for the treatment of diabetic kidney disease.
Article
Full-text available
Diabetic nephropathy (DN) is a chronic inflammatory disease that affects millions of diabetic patients worldwide. The key to treating of DN is early diagnosis and prevention. Once the patient enters the clinical proteinuria stage, renal damage is difficult to reverse. Therefore, developing early treatment methods is critical. DN pathogenesis results from various factors, among which the immune response and inflammation play major roles. Ferroptosis is a newly discovered type of programmed cell death characterized by iron-dependent lipid peroxidation and excessive ROS production. Recent studies have demonstrated that inflammation activation is closely related to the occurrence and development of ferroptosis. Moreover, hyperglycemia induces iron overload, lipid peroxidation, oxidative stress, inflammation, and renal fibrosis, all of which are related to DN pathogenesis, indicating that ferroptosis plays a key role in the development of DN. Therefore, this review focuses on the regulatory mechanisms of ferroptosis, and the mutual regulatory processes involved in the occurrence and development of DN and inflammation. By discussing and analyzing the relationship between ferroptosis and inflammation in the occurrence and development of DN, we can deepen our understanding of DN pathogenesis and develop new therapeutics targeting ferroptosis or inflammation-related regulatory mechanisms for patients with DN.
Article
The pathogenesis of diabetic kidney disease (DKD) is complex, and the existing treatment methods cannot control disease progression well. Macrophages play an important role in the development of DKD. This study aimed to search for biomarkers involved in immune injury induced by macrophages in DKD. The GSE96804 dataset was downloaded and analyzed by the CIBERSORT algorithm to understand the differential infiltration of macrophages between DKD and normal controls. Weighted gene co-expression network analysis was used to explore the correlation between gene expression modules and macrophages in renal tissue of DKD patients. Protein-protein interaction network and machine learning algorithm were used to screen the hub genes in the key modules. Subsequently, the GSE30528 dataset was used to further validate the expression of hub genes and analyze the diagnostic effect by the receiver operating characteristic curve. The clinical data were applied to explore the prognostic significance of hub genes. CIBERSORT analysis showed that macrophages increased significantly in DKD renal tissue samples. A total of ten modules were generated by weighted gene co-expression network analysis, of which the blue module was closely associated with macrophages. The blue module mainly played an important role in biological processes such as immune response and fibrosis. Fibronectin 1 (FN1) and transforming growth factor beta induced (TGFBI) were identified as hub genes of DKD patients. Receiver operating characteristic curve analysis was performed in the test cohort: FN1 and TGFBI had larger area under the curve values (0.99 and 0.88, respectively). Clinical validation showed that 2 hub genes were negatively correlated with the estimated glomerular filtration rate in DKD patients. In addition, FN1 and TGFBI showed a strong positive correlation with macrophage alternative activation. FN1 and TGFBI are promising biomarkers for the diagnosis and treatment of DKD patients, which may participate in immune response and fibrosis induced by macrophages.
Article
Renal fibrosis is a common pathological manifestation in various chronic kidney diseases. Inflammation plays a central role in renal fibrosis development. Owing to their significant participation in inflammation and autoimmunity, chemokines have always been the hot spot and focus of scientific research and clinical intervention. Among the chemokines, monocyte chemoattractant protein-1 (MCP-1), also known as C-C motif chemokine ligand 2, together with its main receptor C–C chemokine receptor type 2 (CCR2) are important chemokines in renal fibrosis. The MCP-1/CCR2 axis is activated when MCP-1 binds to CCR2. Activation of MCP-1/CCR2 axis can induce chemotaxis and activation of inflammatory cells, and initiate a series of signaling cascades in renal fibrosis. It mediates and promotes renal fibrosis by recruiting monocyte, promoting the activation and transdifferentiation of macrophages. This review summarizes the complex physical processes of MCP-1/CCR2 axis in renal fibrosis and addresses its general mechanism in renal fibrosis by using specific examples, together with the progress of targeting MCP-1/CCR2 in renal fibrosis with a view to providing a new direction for renal fibrosis treatment.
Article
Full-text available
Diabetic kidney disease (DKD) is a major cause of end-stage renal disease and imposes a heavy global economic burden; however, little is known about its complicated pathophysiology. Investigating the cellular crosstalk involved in DKD is a promising avenue for gaining a better understanding of its pathogenesis. Nonetheless, the cellular crosstalk of podocytes and endothelial cells in DKD is better understood than that of mesangial cells (MCs) and renal tubular epithelial cells (TECs). As the significance of MCs and TECs in DKD pathophysiology has recently become more apparent, we reviewed the existing literature on the cellular crosstalk of MCs and TECs in the context of DKD to acquire a comprehensive understanding of their cellular communication. Insights into the complicated mechanisms underlying the pathophysiology of DKD would improve its early detection, care, and prognosis.
Article
Full-text available
Understanding functional interactions between G protein-coupled receptors is of great physiological and pathophysiological importance. Heteromerization provides one important potential mechanism for such interaction between different signalling pathways via macromolecular complex formation. Previous studies suggested a functional interplay between angiotensin II receptor type 1 (AT1) and Chemokine (C-C motif) Receptor 2 (CCR2). However the molecular mechanisms are not understood. We investigated AT1-CCR2 functional interaction in vitro using bioluminescence resonance energy transfer in HEK293 cells and in vivo using subtotal-nephrectomized rats as a well-established model for chronic kidney disease. Our data revealed functional heteromers of these receptors resulting in CCR2-Gαi1 coupling being sensitive to AT1 activation, as well as apparent enhanced β-arrestin2 recruitment with agonist co-stimulation that is synergistically reversed by combined antagonist treatment. Moreover, we present in vivo findings where combined treatment with AT1- and CCR2-selective inhibitors was synergistically beneficial in terms of decreasing proteinuria, reducing podocyte loss and preventing renal injury independent of blood pressure in the subtotal-nephrectomized rat model. Our findings further support a role for G protein-coupled receptor functional heteromerization in pathophysiology and provide insights into previous observations indicating the importance of AT1-CCR2 functional interaction in inflammation, renal and hypertensive disorders.
Article
Full-text available
Background Combination therapy with angiotensin-converting–enzyme (ACE) inhibitors and angiotensin-receptor blockers (ARBs) decreases proteinuria; however, its safety and effect on the progression of kidney disease are uncertain. Methods We provided losartan (at a dose of 100 mg per day) to patients with type 2 diabetes, a urinary albumin-to-creatinine ratio (with albumin measured in milligrams and creatinine measured in grams) of at least 300, and an estimated glomerular filtration rate (GFR) of 30.0 to 89.9 ml per minute per 1.73 m² of body-surface area and then randomly assigned them to receive lisinopril (at a dose of 10 to 40 mg per day) or placebo. The primary end point was the first occurrence of a change in the estimated GFR (a decline of ≥30 ml per minute per 1.73 m² if the initial estimated GFR was ≥60 ml per minute per 1.73 m² or a decline of ≥50% if the initial estimated GFR was <60 ml per minute per 1.73 m²), end-stage renal disease (ESRD), or death. The secondary renal end point was the first occurrence of a decline in the estimated GFR or ESRD. Safety outcomes included mortality, hyperkalemia, and acute kidney injury. Results The study was stopped early owing to safety concerns. Among 1448 randomly assigned patients with a median follow-up of 2.2 years, there were 152 primary end-point events in the monotherapy group and 132 in the combination-therapy group (hazard ratio with combination therapy, 0.88; 95% confidence interval [CI], 0.70 to 1.12; P=0.30). A trend toward a benefit from combination therapy with respect to the secondary end point (hazard ratio, 0.78; 95% CI, 0.58 to 1.05; P=0.10) decreased with time (P=0.02 for nonproportionality). There was no benefit with respect to mortality (hazard ratio for death, 1.04; 95% CI, 0.73 to 1.49; P=0.75) or cardiovascular events. Combination therapy increased the risk of hyperkalemia (6.3 events per 100 person-years, vs. 2.6 events per 100 person-years with monotherapy; P<0.001) and acute kidney injury (12.2 vs. 6.7 events per 100 person-years, P<0.001). Conclusions Combination therapy with an ACE inhibitor and an ARB was associated with an increased risk of adverse events among patients with diabetic nephropathy. (Funded by the Cooperative Studies Program of the Department of Veterans Affairs Office of Research and Development; VA NEPHRON-D ClinicalTrials.gov number, NCT00555217.)
Article
Full-text available
CCL2/C-C chemokine receptor 2 (CCR2) signalling is suggested to play a significant role in various kidney diseases including diabetic nephropathy. We investigated the renoprotective effect of a CCR2 antagonist, RS102895, on the development of diabetic nephropathy in a type 2 diabetic mouse model. Six-week-old diabetic db/db and non-diabetic db/m mice were fed either normal chow or chow mixed with 2 mg/kg/day of RS102895 for 9 weeks. We investigated the effects of CCR2 antagonism on blood glucose, blood pressure, albuminuria and the structure and ultrastructure of the kidney. Diabetes-induced albuminuria was significantly improved after CCR2 antagonist treatment, and glucose intolerance was improved in the RS102895-treated diabetic mice. RS102895 did not affect blood pressure, body weight or kidney weight. Mesangial expansion, glomerular basement membrane thickening and increased desmin staining in the diabetic kidney were significantly improved after RS102895 treatment. The up-regulation of vascular endothelial growth factor mRNA expression and the down-regulation of nephrin mRNA expression were markedly improved in the kidneys of RS102895-treated diabetic mice. Increased renal CD68 and arginase II and urinary malondialdehyde in diabetes were effectively attenuated by RS102895 treatment. Blockade of CCL2/CCR2 signalling by RS102895 ameliorates diabetic nephropathy not only by improving blood glucose levels but also by preventing CCL2/CCR2 signalling from altering renal nephrin and VEGF expressions through blocking macrophage infiltration, inflammation and oxidative stress in type 2 diabetic mice.
Article
Full-text available
Background: This study was undertaken to determine whether use of the direct renin inhibitor aliskiren would reduce cardiovascular and renal events in patients with type 2 diabetes and chronic kidney disease, cardiovascular disease, or both. Methods: In a double-blind fashion, we randomly assigned 8561 patients to aliskiren (300 mg daily) or placebo as an adjunct to an angiotensin-converting-enzyme inhibitor or an angiotensin-receptor blocker. The primary end point was a composite of the time to cardiovascular death or a first occurrence of cardiac arrest with resuscitation; nonfatal myocardial infarction; nonfatal stroke; unplanned hospitalization for heart failure; end-stage renal disease, death attributable to kidney failure, or the need for renal-replacement therapy with no dialysis or transplantation available or initiated; or doubling of the baseline serum creatinine level. Results: The trial was stopped prematurely after the second interim efficacy analysis. After a median follow-up of 32.9 months, the primary end point had occurred in 783 patients (18.3%) assigned to aliskiren as compared with 732 (17.1%) assigned to placebo (hazard ratio, 1.08; 95% confidence interval [CI], 0.98 to 1.20; P=0.12). Effects on secondary renal end points were similar. Systolic and diastolic blood pressures were lower with aliskiren (between-group differences, 1.3 and 0.6 mm Hg, respectively) and the mean reduction in the urinary albumin-to-creatinine ratio was greater (between-group difference, 14 percentage points; 95% CI, 11 to 17). The proportion of patients with hyperkalemia (serum potassium level, ≥6 mmol per liter) was significantly higher in the aliskiren group than in the placebo group (11.2% vs. 7.2%), as was the proportion with reported hypotension (12.1% vs. 8.3%) (P<0.001 for both comparisons). Conclusions: The addition of aliskiren to standard therapy with renin-angiotensin system blockade in patients with type 2 diabetes who are at high risk for cardiovascular and renal events is not supported by these data and may even be harmful. (Funded by Novartis; ALTITUDE ClinicalTrials.gov number, NCT00549757.).
Article
In controlled clinical trials, the treatments are likely to be influenced by various prognostic factors, and while assigning treatments sequentially to the patients it is desirable to allot the treatments in such a way that the treatments are balanced over the main effects of prognostic factors and also on some or all interactions between the prognostic factors if the interactions are present. Efran (1971), Pocock and Simon (1975) and Freedman and White (1976) described some methods of balancing the treatments over the prognostic factors. In this paper, we shall describe a new approach in assigning the treatments using multivariate methods.
Article
Study background: Inflammatory macrophages expressing the C-C chemokine receptor 2 (CCR2) accumulate in adipose tissue and contribute to insulin resistance. CCX140-B is an orally-administered antagonist of CCR2 expressed on monocytes and macrophages and blocks infiltration of these cells into adipose tissue. A pilot Phase 2 clinical trial was conducted in patients with type 2 diabetes with the primary objective to evaluate the safety and tolerability of CCX140-B. Key secondary objectives included assessment of glycemic parameters, fasting plasma glucose (FPG) and hemoglobin A1c (HbA1c). Methods: This is a randomized, double-blind, clinical trial of CCX140-B in 159 subjects with type 2 diabetes on stable metformin for at least 8 weeks prior to study entry. HbA 1c was 6.5 to 10% and FPG 135 to 270 mg/dL at study entry. Randomized subjects received double-blind placebo (N=32), 5 mg CCX140-B (N=63), 10 mg CCX140-B (N=32), or pioglitazone 30 mg (N=32) once daily orally for 4 weeks, with a 4-week follow-up period. Results: CCX140-B was well tolerated. No serious adverse events occurred with CCX140-B. FPG showed a CCX140-B dose-dependent decrease, with 10 mg CCX140-B showing a similar decrease to pioglitazone (leastsquares mean change at week 4 of -16.1 vs. -21.4 mg/dL, respectively). HbA 1c least-squares mean changes from baseline to week 4 for the placebo, 5 mg CCX140-B, 10 mg CCX140-B, and pioglitazone groups were -0.09%, -0.09%, -0.23% (p=0.045 vs. placebo), and -0.13% (NS vs. placebo), respectively. No detrimental changes were seen in plasma monocyte chemoattractant protein-1 or blood monocyte counts with CCX140-B. Conclusion: CCX140-B, an orally administered, specific CCR2 antagonist was found to be well tolerated and safe in this Phase 2 clinical trial and showed evidence of a beneficial effect on glycemic parameters. If confirmed, CCX140-B might be beneficial in treatment of patients with type 2 diabetes. (Clinical Trial Registry: clinicaltrials.gov identifier NCT01028963).
Article
Albuminuria has been proposed as a surrogate end point in randomized clinical trials of renal disease progression. Most evidence comes from observational analyses showing that treatment-induced short-term changes in albuminuria correlate with risk change for ESRD. However, such studies are prone to selection bias and residual confounding. To minimize this bias, we performed a meta-analysis of clinical trials to correlate the placebo-corrected drug effect on albuminuria and ESRD to more reliably delineate the association between changes in albuminuria and ESRD. MEDLINE and EMBASE were searched for clinical trials reported between 1950 and April 2014. Included trials had a mean follow-up of ≥1000 patient-years, reported ESRD outcomes, and measured albuminuria at baseline and during follow-up. Twenty-one clinical trials involving 78,342 patients and 4183 ESRD events were included. Median time to first albuminuria measurement was 6 months. Fourteen trials tested the effect of renin-angiotensin-aldosterone-system inhibitors and seven trials tested other interventions. We observed variability across trials in the treatment effect on albuminuria (range, -1.3% to -32.1%) and ESRD (range, -55% to +35% risk change). Meta-regression analysis revealed that the placebo-adjusted treatment effect on albuminuria significantly correlated with the treatment effect on ESRD: for each 30% reduction in albuminuria, the risk of ESRD decreased by 23.7% (95% confidence interval, 11.4% to 34.2%; P=0.001). The association was consistent regardless of drug class (P=0.73) or other patient or trial characteristics. These findings suggest albuminuria may be a valid substitute for ESRD in many circumstances, even taking into account possible other drug-specific effects that may alter renal outcomes. Copyright © 2014 by the American Society of Nephrology.