Skip to main content
Erschienen in: BMC Nephrology 1/2015

Open Access 01.12.2015 | Research article

Orally active prostacyclin analogue beraprost sodium in patients with chronic kidney disease: a randomized, double-blind, placebo-controlled, phase II dose finding trial

verfasst von: Akio Koyama, Toshiro Fujita, Fumitake Gejyo, Hideki Origasa, Masanao Isono, Hajimu Kurumatani, Kiyonobu Okada, Hiroyuki Kanoh, Takashi Kiriyama, Shunsuke Yamada

Erschienen in: BMC Nephrology | Ausgabe 1/2015

Abstract

Background

Evidence increasingly points to the importance of chronic hypoxia in the tubulointerstitium as a final common pathway to end-stage renal disease (ESRD). Beraprost sodium (BPS) is an orally active prostacyclin (PGI2) analogue demonstrating prevention of the progression of chronic kidney disease (CKD) in various animal models by maintaining renal blood flow and attenuating renal ischemic condition.

Methods

This multicenter, randomized, double-blind, placebo-controlled, phase II trial was designed to determine the recommended dose of the sustained-release form of BPS (TRK-100STP 120 μg/day or 240 μg/day) in Japanese patients with CKD. TRK-100STP was administered to a total of 112 patients. The primary efficacy endpoint was the difference in the slope of the regression line of reciprocal of serum creatinine (1/SCr) over time, obtained by the least-squares method.

Results

Regarding the primary endpoint, statistical superiority of TRK-100STP 240 μg over placebo was not confirmed and so a recommended dose was not determined. Compared to placebo, however, the slope of regression line of 1/SCr, elevation of SCr and serum cystatin C during the treatment period revealed greater improvement at 120 μg, at both doses, and at 240 μg, respectively. In terms of safety, both TRK-100STP treatment groups were well tolerated.

Conclusions

Although the study failed to meet the primary endpoint, results indicate that TRK-100STP may potentially prevent the decline in renal function of CKD patients independent of blood pressure or urinary protein levels.

Trial registration

NCT02480751. June 21, 2015.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s12882-015-0130-5) contains supplementary material, which is available to authorized users.

Competing interests

AK and FG have no conflict of interest. TF and HO serve as a consultant of Toray. TF is a recipient of grants for Astellas, Toray, Boehringer Ingelheim, Chugai, Fukuda Denshi, Kyowa Hakko Kirin, Mitsubishi Tanabe, Mochida, Omron, Pfizer and Takeda. Also, TF is a recipient of lecture fee for Astellas, Boehringer Ingelheim, Kyowa Hakko Kirin, Mitsubishi Tanabe, Daiichi Sankyo, Novartis, Pfizer and Takeda. MI, HAKU and KO are employees of Toray, while HIKA, TK and SY are employees of Astellas.

Authors’ contributions

AK, TF and FG were assigned as medical advisors for the study but their terms of duty were all different during the study period. They were involved in the conception and design of the study, and advisers for conducting the study. HO was involved in the conception and study design and provided statistical support of the study. MI, HAKU, KO, HIKA, TK and SY were involved in the study conception and acquisition of data and data analysis. AK and HAKU wrote the draft of the manuscript. All authors critically reviewed the manuscript and approved the final manuscript as submitted.
Abkürzungen
ACEIs
Angiotensin-converting enzyme inhibitors
ADR
Adverse drug reaction
AE
Adverse event
ANCOVA
Analysis of covariance
ARB
Angiotensin-II receptor blocker
BPS
Beraprost sodium
CASSIOPEIR
CRF Asian Study with Oral PGI2 derivative for Evaluating Improvement of Renal function
CCr
Creatinine clearance
CKD
Chronic kidney disease
CRF
Chronic renal failure
ECG
Electrocardiogram
ESRD
End-stage renal disease
FAS
Full analysis set
NSAIDs
Nonsteroidal anti-inflammatory drugs
PGI2
Prostaglandin I2 (also called prostacyclin)
PPS
Per protocol set
RAS
Renin-angiotensin system
SADR
Serious adverse drug reactions
SCr
Serum creatinine

Background

An increasing number of patients with end-stage renal disease (ESRD) require dialysis or transplantation. Although diabetic nephropathy is a major reason for eventual ESRD, primary glomerular diseases and nephrosclerosis still comprise significant proportions of chronic kidney disease (CKD) patients, especially in Asian countries [14].
Angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin-II receptor blockers (ARBs) are often administered to both diabetic and non-diabetic nephropathy patients, and are established as recommended treatment agents for non-diabetic nephropathy patients with albumin excretion [5]. However, it is evident that these agents are insufficient for the prevention of progressive renal disease. In addition, the use of combination therapy of renin-angiotensin system (RAS) inhibitors such as ACEI and ARB has demonstrated in some recent clinical trials to be not effective [69]. These findings suggest an urgent clinical need for a new treatment option able to significantly delay the progression of CKD.
Prostacyclin (PGI2) is primarily synthesized in endothelial cells and one of the important functions is to protect kidneys from ischemic damage in pathophysiological conditions [10], as evidenced, for example, by the development of renal impairment in prostacyclin synthase knockout mice [11]. Attempts have been made to use PGI2 or its analogues in patients with kidney disease; the PGI2 analogue iloprost was successfully used for patients with contrast media-induced nephropathy [12].
Beraprost sodium (BPS) is an orally active PGI2 analogue [13] and TRK-100STP is its sustained-release form [14]; both were generated and developed by Toray Industries, Inc. In Asian countries, the immediate-release form of BPS has been widely used in the treatment of patients with chronic arterial occlusion and pulmonary arterial hypertension [15].
Recent evidence increasingly points to the importance of chronic hypoxia especially in the tubulointerstitium as a final common pathway to the progression of CKD [1620]. BPS prevents the progression of CKD in various animal models [2124] by maintaining renal microvasculature and blood flow [25, 26]. In addition, it is reported that BPS is effective in Acute Kidney Injury such as contrast nephropathy and cisplatin nephropathy [27, 28]. These effects of BPS are thought to be based on multiple mechanisms of actions: direct protective effect on vascular endothelial cells [29]; inhibition of the production of inflammatory cytokines from monocyte/macrophages [23]; vasodilative [30] and antiplatelet effects [31]. The effects of BPS have also been assessed in two studies in patients with glomerulonephritis [32, 33]. Although these studies were open-label and did not have control group, the results of one study suggested that BPS mitigates the progression rate of renal dysfunction [32, 33] by increasing renal blood flow without glomerular hyperfiltration.
The efficacy of BPS on diabetic nephropathy has also been reported in several clinical [34] and non-clinical studies [3537]; however, several methodological limitation are evident with regards to the involvement of both diabetic and non-diabetic CKD patients in a single protocol. Given the evident unmet need for non-diabetic nephropathy treatment in Asia and considering that much of the non-clinical data on BPS has been generated in non-diabetic CKD, the priority and focus of the present study was on non-diabetic CKD, given that renal disorders are not modified by diabetes. This is the first randomized, placebo-controlled, double-blind, comparative trial to investigate the recommended dose at which TRK-100STP suppresses the progression of CKD in patients with primary glomerular disease or nephrosclerosis.

Methods

This multicenter, randomized, double-blind placebo-controlled, phase II trial was designed to determine the recommended dose of TRK-100STP (i.e., either 120 μg/day or 240 μg/day) for suppressing progression in Japanese patients with primary glomerular disease or nephrosclerosis. The study was approved by each participating center’s local Research Ethics Committee. Only patients with written informed consent were included (Please refer to the list of participating centers at the end of the paper).

Study design

A summary of the study method is presented in Fig. 1. After the run-in period, when placebo tablets were orally administered twice daily for 22 weeks in a single-blinded manner, patients who met all of the inclusion criteria and none of the exclusion criteria (detailed in Fig. 2) were randomized into one of the three treatment groups (TRK-100STP 120 μg, 240 μg or placebo group) using computer random number generator to select random permuted blocks. Central randomization was performed by external statistician for the 28-week treatment period in a double-blinded manner. Enrollment to the trial was restricted to patients whose renal function had progressively declined during the 22-week run-in period (R0 ~ R20) to obtain adequate treatment response data during the 28-week treatment period (W0 ~ W28).
Reciprocal of serum creatinine (1/SCr) versus time slope shows the speed of renal function decline so that this parameter is used to estimate how long it will take to start dialysis and to evaluate therapeutic effects [38]. This parameter is used for example to show the status of nephropathy in the RENAAL trial [39].
Therefore, in order to limit the participants to those with progressive diseases, only patients whose reciprocal of serum creatinine (1/SCr) versus time slope was ≤ −0.005 dL/mg over 4 weeks during the run-in period were randomized. This value was based on the patients’ background in a trial of orally administered spherical carbon adsorbent AST-120 [40]. The primary endpoint was also 1/SCr versus time slope and the differences between the treatment- and the observation-period values were evaluated.
The study drug in the treatment period was orally administered twice daily, after meals, for 28 weeks in a double-blinded manner. The first 2 weeks of the treatment period were designated as the dose-titration period with respect to safety analyses. The initial dose for the TRK-100STP groups was 120 μg/day. For the 240 μg group, the dose was increased to 240 μg/day after the 2-week dose-titration period. One follow-up assessment was performed between 7 to 35 days after completion or discontinuation of study drug during the treatment period.
The two doses of TRK-100STP (120 μg/day and 240 μg/day) were selected for this trial on the basis that a 30 μg single dose was considered unable to achieve the effective plasma concentration, whilst a 180 μg single dose previously resulted in a higher incidence of headache and other adverse drug reactions (ADRs) in a completed phase I clinical study of TRK-100STP. Based on these observations, TRK-100STP at 60 to 120 μg b.i.d. (i.e., 120 to 240 μg/day) were selected for investigation in the present trial.

Study endpoints

The primary efficacy endpoint was the difference between the run-in and treatment periods in the slope of the regression line of 1/SCr versus time, which was calculated by the least-squares method. All SCr data collected in the run-in period (R0 ~ R20) and the treatment period (W4 ~ W28) were used for the plot in order to exclude the possible effect of the titration period (W0 ~ W2).
Secondary endpoints were as follows: 1) The difference between the run-in period (R0 ~ R20) and treatment period (W0 ~ W28) in 1/SCr versus time slope; 2) changes in creatinine clearance (CCr, Cockcroft-Gault equation); 3) changes in CCr (urinalysis with 24-hour pooled urine); 4) changes in urinary protein excretion; 5) ratio of SCr; and 6) changes in serum cystatin C. Safety endpoints were as follows: 1) adverse events (AEs); 2) clinical laboratory tests; 3) vital signs and body weight; and 4) 12-lead electrocardiogram (ECG).

Prohibited and restricted concomitant medications

Prohibited concomitant medications

Use of the following medications were prohibited during the study period: spherical carbon adsorbent; prostaglandin analogues, other than eye drops and those in ointment form; antiplatelet agents, with the exception of aspirin products; anticoagulant and thrombolytic agents, except for temporary use such as for examinations; fluorinated pyrimidine antifungal agents; and iodinated radiocontrast agents.

Restricted concomitant medications

Dosage of ACEIs and ARBs were stipulated to be fixed during the study period. However, in necessary situations, such as increased serum potassium levels that did not improve with alternative options, a reduction in ACEI or ARB doses was allowed. Taking nonsteroidal anti-inflammatory drugs (NSAIDs) continuously for more than one week was prohibited.

Sample size estimation

The sample size estimation was based on a previous study, a Phase III clinical trial of AST-120 (indicated for the treatment of chronic renal failure (CRF) in Japan), conducted in a total of 237 patients (119 patients treated with the active drug and 118 received placebo) for 24 weeks [40]. The study showed that the difference in the slope of regression line of 1/SCr versus time of AST-120 and the placebo groups before and after treatment with the test drug was 0.00352 (dL/mg over 4 weeks). Based on this value, we hypothesized that the TRK-100STP 120 μg group would show efficacy similar to the AST-120 group, and that the TRK-100STP 240 μg group would show 1.5 times better efficacy than the AST-120 group. According to these predictions, we set contrast coefficients [−1, 0, 1] for the placebo, the TRK-100STP 120 μg, and the 240 μg groups. The number of patients required per group was estimated to be 71 (with a two-sided 5 % significance level and 80 % statistical power). On this basis, the target number of patients needed to recruit was set at 430 patients, taking into account the possibility that the patient withdrawal rate in the run-in period might be high because of the long run-in period.

Statistical analysis

(a) Primary endpoint
Using all SCr data collected during the run-in period (R0 ~ R20) and treatment period (W4 ~ W28), reciprocal SCr values plotted against time were analyzed. The slopes of the regression line were calculated by applying the least-squares equation. The difference in the slopes between the run-in period (R0 ~ R20) and treatment period (W4 ~ W28) was evaluated as the primary endpoint. In order to determine the recommended dose, the primary endpoint was analyzed by the following procedures:
  • Step 1: Analysis of covariance (ANCOVA) with the slopes calculated from the results obtained during the run-in period as a covariate after assigning contrast coefficients [−1, 0, 1].
  • Step 2: ANCOVA with the slopes obtained from the results collected during the run-in period as a covariate after assigning contrast coefficient [−1, 1, 0], only when a statistical significance was detected in Step 1.
(b) Secondary endpoints
Analytical methods for secondary endpoints are described in each result.
(c) Post hoc analysis
Ratio of SCr (the final evaluation point/W0)
In analyzing the ratio of SCr for the secondary endpoint, the SCr values measured at R20 were chosen as baseline as other parameters such as CCr, urinary protein and cystatin C were measured only at R20. Regarding SCr, however, as the value at W0 measured just before the treatment period was available, the ratio of SCr (the final evaluation point/W0) was assessed by ANCOVA with the SCr (R20) as covariate.
Change in serum cystatin C in the treatment period (final evaluation point - R20)
As the ratio of SCr was analyzed only for the treatment period, changes in cystatin C were re-evaluated based only on data from the treatment period.
Changes in cystatin C were calculated using values taken at R20 as the baseline (the final evaluation point – R20). ANCOVA was performed with the SCr (R20) as the covariate. As the values at W0 were not available, the values at R20, the examination point of the study closest to W0, were used as baseline.
Ratio of eGFR (the final evaluation point/W0)
Ratio of eGFR (the final evaluation point/W0) was assessed by ANCOVA with the SCr (R20) as covariate. In order to calculate the eGFR for Japanese patients, the following equation was used;
GFR(male) = 194*Scr-1.094*age-0.287, and GFR(female) = GFR(male)*0.739 [41].

Study measurements

The parameters that were measured and analyzed are listed in Table 1.
Table 1
Study measurements
Examination by investigator
Objective symptoms (patient interview)
Physical examination
Body weight, blood pressure, pulse rate, 12-lead ECG
Hematological examination
WBC, RBC, Hb, Ht, PLT,
Differential count of leukocytes (basophil, eosinophil, neutrophil, lymphocyte, monocyte)
Bleeding and coagulation test
PT, APTT
Blood biochemistry
TP, Alb, T-BIL, AST(GOT), ALT(GPT), ALP, LDH, γ-GTP,
TCh, TG, UA, BUN, serum creatinine (SCr), Na, K, Cl, Ca, P, HbA1c, cystatin C,
High-sensitive CRP
Uremia toxins test in plasma
Guanidino succinate
Urinalysis (occasional urine)
pH, qualitative protein, glucose, and urobilinogen, occult blood reaction urinary sediment (RBC, WBC, casts), β2 microglobulin (adjusted by creatinine), pregnancy test (hCG)
Urinalysis (24-hour pooled urine)
Urinary protein excretion, CCr, urea nitrogen, electrolytes (Na, Cl), urine output, creatinine

Trial registration

NCT02480751. June 21, 2015.

Results

Patient disposition

The trial began on October 24, 2005 (the date the first consent was obtained) and was completed on May 20, 2008 (the last follow-up date of the final patient).
A summary of the patient disposition is described in Fig. 3. Written informed consents were obtained from a total of 431 patients, 11 of whom did not meet the criteria on pre-registration and a total of 420 patients were pre-registered. Of these 420 patients, 29 dropped out before the start of the run-in period and a further 278 patients were withdrawn during the run-in period or at the registration. A total of 113 patients were therefore registered, of which one patient, who mistakenly took the run-in period medication and did not take the study drug for the treatment period after the registration, was excluded from the study. As a result, the study drugs were administered to a final total of 112 patients (35 patients in the placebo group, 36 patients in the 120 μg group and 41 patients in the 240 μg group) in a double-blind manner. Most of 238 patients who did not meet the inclusion/exclusion criteria before registration were excluded because their renal failure progressed at a slower rate during the run-in period, represented by a 1/SCr versus time slope of ≤ −0.005 dL/mg over 4 weeks.

Analysis sets

Ten patients whose SCr values measured less than three points after Week 4 of the treatment period were excluded from 112 patients randomized. As a result, 102 patients (34 patients in the placebo group, 32 patients in the 120 μg group and 36 patients in the 240 μg group) were included in the full analysis set (FAS). From FAS, four patients were excluded whose SCr values measured after Week 4 of the treatment period were less than 5 points. The remaining patients constituted the per protocol set (PPS). PPS therefore consisted of 98 patients (34 patients in the placebo group, 29 patients in the 120 μg group and 35 patients in the 240 μg group). The safety analysis set consisted of 112 patients (35 patients in the placebo group, 36 patients in the 120 μg group and 41 patients in the 240 μg group), who received the study drug for the treatment period.
Demographic and other baseline characteristics of patients in the FAS are presented in Table 2. Baseline characteristics of patients were not significantly different among the treatment groups. It is noteworthy that 69 % to 81 % of patients were taking ARB in each group.
Table 2
Baseline characteristics of patients in the full analysis set (FAS)
Parameter
Treatment group
Placebo
120 μg
240 μg
(n = 34)
(n = 32)
(n = 36)
Sex
Male
18 (52.9 %)
17 (53.1 %)
24 (66.7 %)
Female
16 (47.1 %)
15 (46.9 %)
12 (33.3 %)
Age
Mean ± SD (years)
59.9 ± 10.0
56.5 ± 14.7
57.8 ± 13.9
Primary disease
Primary glomerular disease
28 (82.4 %)
27 (84.4 %)
25 (69.4 %)
Nephrosclerosis
6 (17.6 %)
5 (15.6 %)
11 (30.6 %)
1/SCr time slope during the run-in period
Mean ± SD (dL/mg over 4 weeks)
−0.01210 ± 0.00497
−0.01535 ± 0.00808
−0.01198 ± 0.00788
SCra
Mean ± SD (mg/dL)
2.377 ± 0.665
2.251 ± 0.618
2.564 ± 0.705
Urinary protein excretiona
Mean ± SD (mg/day)
2103.7 ± 1523.0
2037.5 ± 1763.4
1753.9 ± 1396.2
Systolic blood pressurea
Mean ± SD (mmHg)
122.7 ± 16.1
129.0 ± 13.3
129.4 ± 15.6
Diastolic blood pressurea
Mean ± SD (mmHg)
72.5 ± 11.0
73.4 ± 10.3
75.1 ± 10.6
Concomitant medication
ACEI (+)
13 (38.2 %)
9 (28.1 %)
12 (33.3 %)
ACEI/ARB positive
ARB (+)
25 (73.5 %)
26 (81.3 %)
25 (69.4 %)
aAs of R20

Changes in SCr

Figure 4 illustrates the changes in SCr through the study periods when SCr values of each group at R20 were set at 100 %. The SCr values in all three groups increased during the run-in and treatment periods, and increases in SCr values in the treatment period tends to be suppressed in the two TRK-100STP groups.

Efficacy

As the primary analysis set is also the FAS, the data and the results of evaluation are described based on the FAS, whilst the efficacy analysis was performed in both the FAS and PPS.
(a) Primary endpoint
The difference in the 1/SCr versus time slope between the run-in period (R0 ~ R20) and the treatment period (W4 ~ W28)
As shown in Table 3 and Fig. 5, compared with the run-in period, a slight improvement in the 1/SCr versus time slope was observed during the treatment period in all treatment groups; this tendency was clearly observed in patients of the TRK-100STP 120 μg group.
Table 3
Summary statistics of primary endpoint (the difference in the 1/SCr versus time slope between run-in period [R0 ~ R20] and treatment period [W4 ~ W28])
Treatment group
No. of patients
1/SCr versus time slope (dL/mg over 4 weeks)
Difference in the 1/SCr versus time slope (dL/mg over 4 weeks)
  
Run-in period (R0 ~ R20)
Treatment period (W4 ~ W28)
  
  
Mean
SD
Mean
SD
Mean
SD
Placebo
34
−0.0121
0.00497
−0.0074
0.00935
0.0047
0.01087
120 μg
32
−0.0154
0.00808
−0.0040
0.01345
0.0113
0.01011
240 μg
36
−0.0120
0.00788
−0.0045
0.00750
0.0075
0.00953
Table 4 shows the result of ANCOVA with the 1/SCr versus time slope during the run-in period as the covariate. No statistically significant difference was observed between the 240 μg group and the placebo group when the ANCOVA was performed with the contrast coefficients of [−1, 0, 1] for [the placebo group, the 120 μg group, and the 240 μg group] (P = 0.2234).
Table 4
Difference in the 1/SCr versus time slope between run-in period (R0 ~ R20), and Treatment period (W4 ~ W28) analyzed by the ANCOVA model: comparison of the TRK-100STP groups versus placebo
Treatment group
No. of patients
Least square mean of the change in the 1/SCr slope
Difference in the least square mean (versus placebo)a
Contrast test
(P value)
Point estimate
95 % CI
Point estimate
95 % CI
(−1,0,1)
Placebo
34
0.00516
[0.00187, 0.00845]
 
120 μg
32
0.01026
[0.00682, 0.01369]
0.00510
[0.00031, 0.00989]
 
240 μg
36
0.00798
[0.00478, 0.01118]
0.00282
[−0.00175, 0.00740]
0.2234
a(120 μg group or 240 μg group) – (placebo group)
1/Scr versus time slope during the run-in period as the covariate
(b) Secondary endpoints
The difference in the 1/SCr versus time slope between the run-in period (R0 ~ R20) and the treatment period (W0 ~ W28)
The least square mean of the difference in the 1/SCr versus time slope [95 % CI] was 0.00590 dL/mg over 4 weeks [0.00308, 0.00873] in the placebo group, 0.01065 dL/mg over 4 weeks [0.00769, 0.01360] in the TRK-100STP 120 μg group and 0.00801 dL/mg over 4 weeks [0.00525, 0.01076] in the 240 μg group. Moreover, ANCOVA was performed on the same model as the primary endpoint with the contrast coefficients of [−1, 0, 1], [−1, 1, 0], [−1, −1, 2], and [−2, 1, 1] for the placebo group, the 120 μg group, and the 240 μg group, respectively. The results of ANCOVA with contrast coefficients of [−1, 0, 1], [−1, 1, 0], [−1, −1, 2] and [−2, 1, 1] were P = 0.2912, P = 0.0244, P = 0.8764 and P = 0.0534, respectively. Thus the 120 μg group showed a significant amelioration compared with the placebo group (contrast coefficients: [−1, 1, 0]: P = 0.0244).
Changes in CCr
The difference in the change of CCr in urinalysis with 24-hour pooled urine sample (the change in the treatment period [the final evaluation point – R20]) was 1.08 ± 9.35 mL/min in the placebo group (mean ± SD), 6.16 ± 11.22 mL/min in the TRK-100STP 120 μg group and 3.94 ± 7.80 mL/min in the 240 μg group. Both TRK-100STP groups therefore showed a tendency of inhibiting the decline in CCr as compared with the placebo group (P = 0.0551, P = 0.1800: t-test). When CCr was calculated with the Cockcroft-Gault equation, the change in the treatment period (the final evaluation point – R20) was 2.03 ± 5.98 mL/min in the placebo group (mean ± SD), 3.93 ± 5.30 mL/min in the TRK-100STP 120 μg group and 3.77 ± 5.06 mL/min in the 240 μg group. Both TRK-100STP groups therefore showed a tendency of inhibiting the decline in CCr as compared with the placebo group (P = 0.1771, P = 0.1917: t-test).
Changes in urinary protein excretion
The change in urinary protein excretion (the final evaluation point – R20) was 121.59 ± 715.60 mg/day in the placebo group, 241.68 ± 849.23 mg/day in the TRK-100STP 120 μg group and 280.38 ± 817.97 mg/day in 240 μg group. It was shown that the observed change in the urinary protein excretion in the 120 μg and 240 μg groups (versus the placebo group) was small relative to the standard deviation, and both TRK-100STP groups showed no significant difference compared to the placebo group (P = 0.5457, P = 0.4056: t-test).
Ratio of SCr
The ratio of SCr (the final evaluation point/R20) was 1.14 ± 0.27 in the placebo group, 1.06 ± 0.20 in the TRK-100STP 120 μg group and 1.06 ± 0.13 in the 240 μg group. Although both TRK-100STP groups did not show any significant inhibition, they showed a tendency of inhibiting the increase in SCr as compared with the placebo group (P = 0.1379, P = 0.0942: t-test).
Changes in serum cystatin C
The difference in the change in serum cystatin C (the change in the treatment period [the final evaluation point – R20] – the change in the Run-in period [R20 – R0]) was 0.155 ± 0.448 mg/L in the placebo group, 0.006 ± 0.377 mg/L in the TRK-100STP 120 μg group, and −0.023 ± 0.645 mg/L in the 240 μg group. Both TRK-100STP groups therefore showed a tendency of inhibiting the increase in serum cystatin C as compared versus the placebo group (P = 0.1588, P = 0.1973: t-test).
(c) Post hoc analysis
Ratio of SCr (final evaluation point/W0)
As shown in Table 5 and Fig. 6, both the 120 μg and the 240 μg groups showed a significant inhibition of the increase in SCr ratios as compared with the placebo group.
Table 5
Ratio of SCr [the final evaluation pointa/W0 (week 0 of the treatment period)] analyzed by the ANCOVA model
Treatment group
Number of patients
Least square mean of the ratio of SCr
Difference in the least square mean (versus placebo)
P-value*
Point estimate
SE
Point estimate
SE
Placebo
34
1.169
0.032
120 μg
32
1.069
0.033
0.100
0.045
0.0309
240 μg
36
1.064
0.031
0.105
0.044
0.0204
aAt Week 28 of the treatment period or when treatment was discontinued
*ANCOVA with baseline (SCr(R20)) as covariate
Change in serum cystatin C in treatment period (final evaluation point – R20)
As shown in Table 6 and Fig. 7, the 120 μg group showed a tendency and the 240 μg groups showed significant inhibition of the increase in change in serum cystatin C as compared with the placebo group.
Table 6
Changes in serum cystatin C [the final evaluation pointa-R20 (week 20 of the Run-in period)] comparison of TRK-100STP groups versus placebo analyzed by the ANCOVA model
Treatment group
Number of patientsb
Least square mean of the difference in serum cystatin C (mg/L)
Difference in the least square mean (versus placebo) (mg/L)
P-value
Point estimate
SE
Point estimate
SE
Placebo
32
0.253
0.065
120 μg
31
0.097
0.066
0.157
0.092
0.0928
240 μg
35
0.052
0.063
0.201
0.090
0.0285
aAt Week 28 of the treatment period or when treatment was discontinued
bDifferent from FAS because some patients lacked value of cystatin C

Ratio of eGFR

As shown in Table 7, the 240 μg group showed a tendency and the 120 μg groups showed significant inhibition of the decrease in eGFR ratios as compared with the placebo group.
Table 7
Change in eGFR [the final evaluation pointa/W0 (week 0 of the treatment period)] analyzed by the ANCOVA model
Treatment group
Number of patients
Least square mean of the difference in serum cystatin C (mg/L)
Difference in the least square mean (versus placebo) (mg/L)
P-value
Point estimate
SE
Point estimate
SE
Placebo
34
0.877
0.025
120 μg
32
0.952
0.026
0.075
0.036
0.0365
240 μg
36
0.942
0.024
0.065
0.035
0.0628
aAt Week 28 of the treatment period or when treatment was discontinued

Safety and tolerability

The safety analysis was conducted based on any AEs reported during the study period. The incidence of AEs are summarized in Table 8. Two death were observed in this study; they occurred during the run-in period and so any causal relationships with the study drug was excluded.
Table 8
Summary of the incidence of adverse events
Parameter
120 μg
240 μg
Placebo
Number of patients evaluated
36
41
35
Number of patients who experienced adverse events
27 (75.0 %)
36 (87.8 %)
28 (80.0 %)
Number of patients who experienced serious adverse events
3 (8.3 %)
4 (9.8 %)
1 (2.9 %)
Number of patients who discontinued the study treatment due to adverse events
4 (11.1 %)
7 (17.1 %)
0 (0.0 %)
Number of patients who interrupted the study treatment due to adverse events
0 (0.0 %)
2 (4.9 %)
0 (0.0 %)
(): Incidence rate
The incidence of ADRs are summarized in Table 9. All of these ADRs cases were mild to moderate in severity, and recovered without treatment. Two serious adverse drug reactions (SADR) of cardiac failure and acute myocardial infarction were observed in the 120 μg group after randomization and both patients recovered.
Table 9
Summary of adverse drug reaction incidence
Parameter
120 μg
240 μg
Placebo
Number of patients evaluated
36
41
35
Number of patients who experienced ADR
7 (19.4 %)
13 (31.7 %)
5 (14.3 %)
Number of patients who experienced serious ADR
2 (5.6 %)
0 (0.0 %)
0 (0.0 %)
Number of patients who discontinued the study treatment due to ADR
3 (8.3 %)
5 (12.2 %)
0 (0.0 %)
Number of patients who interrupted the study treatment due to ADR
0 (0.0 %)
1 (2.4 %)
0 (0.0 %)
(): Incidence rate
The AEs and ADRs that occurred at an incidence of 5 % or more are summarized in Table 10. The severity of headache in ADR was as follows: moderate headache was reported in 1 patient in the 240 μg group; mild headache was reported in 1 patient in the 120 μg group, and 5 patients in the 240 μg group. All of these patients recovered without treatment. For clinical laboratory tests, vital signs, body weight and 12-lead ECG, no specific concerns were observed. TRK-100STP did not have significant effect on blood pressure at any dose, as detailed in Table 11.
Table 10
Adverse events and adverse drug reactions with an incidence of 5 % or more
Adverse Events
120 μg
240 μg
Placebo
Headache
5 (13.9 %)
Nasopharyngitis
11 (26.8 %)
Nasopharyngitis
8 (22.9 %)
Nasopharyngitis
4 (11.1 %)
Headache
7 (17.1 %)
Pruritus
3 (8.6 %)
Back pain
3 (8.3 %)
Diarrhea
5 (12.2 %)
Fever
3 (8.6 %)
Hyperkalemia
2 (5.6 %)
Malaise
3 (7.3 %)
Hyperkalemia
3 (8.6 %)
Dizziness
2 (5.6 %)
  
Influenza
2 (5.7 %)
Hypertension
2 (5.6 %)
  
Hyperkalemia
2 (5.7 %)
Upper respiratory tract inflammation
2 (5.6 %)
  
Muscle Spasm
2 (5.7 %)
Diarrhea
2 (5.6 %)
  
Genital Bleeding
2 (5.7 %)
Vomiting
2 (5.6 %)
    
Arthralgia
2 (5.6 %)
    
Adverse drug reactions
120 μg
240 μg
Placebo
Hypertension
2 (5.6 %)
Headache
6 (14.6 %)
Genital Bleeding
2 (5.7 %)
  
Malaise
3 (7.3 %)
  
(): Incidence rate
Table 11
Effect of TRK-100STP 120 μg and 240 μg on blood pressure
Treatment group
Systolic blood pressure
Diastolic blood pressure
R20
Final evaluation point
R20
Final evaluation point
Placebo
No. of patients
35
35
35
35
Mean ± SD
123.8 ± 17.0
127.5 ± 20.3
72.7 ± 10.9
75.3 ± 9.9
120 μg
No. of patients
36
35
36
35
Mean ± SD
128.3 ± 13.0
127.8 ± 13.5
73.6 ± 10.7
75.4 ± 10.5
240 μg
No. of patients
41
40
41
40
Mean ± SD
128.3 ± 16.4
129.4 ± 13.7
74.4 ± 10.6
75.1 ± 8.9
(Unit of blood pressure: mmHg)

Discussion

This is the first randomized, double-blind, placebo controlled comparative trial of TRK-100STP to investigate the possible therapeutic dose of TRK-100STP in CKD patients with a primary disease of glomerular disease or nephrosclerosis.
In the FAS, statistical superiority of the 240 μg group over the placebo could not be confirmed by the primary endpoint, and so the putative recommended dose could not be clearly determined. One of the reasons why statistically significant superiority could not be confirmed was the trial lost statistical power due to the fact that more patients than had been expected were withdrawn from the study during the run-in period. As we needed to consider the feasibility of study completion, we could not secure the sufficient number of patients for analysis.
This is considered to be because the pace at which the decline in renal function became slower during the run-in period. As shown in Table 3, 1/SCr versus time slope in the treatment period was ameliorated even in the placebo group. In this trial, the CKD patients who progressed and whose 1/SCr versus time slopes were ≤ −0.005 dL/mg over 4 weeks were selected at the start of the treatment period. As a result, the number of patients to be enrolled was reduced to about 1/3 of patients who had been screened. It is thought that such selection of patients also led to the regression toward the mean in all groups. This amelioration in the 1/SCr versus time slope in the placebo group may have made it difficult to detect the difference between the placebo group and the 240 μg group in terms of the primary endpoint. However, our trial showed, for the first time, that TRK-100STP might prevent the decline of renal function. This conclusion was supported by the following facts: both active study drug treatment groups exhibited better efficacy than the placebo group, and the efficacy was particularly high in the 120 μg group on the primary endpoint. In the secondary endpoint analysis, both TRK-100STP groups showed improvements on each of the renal filtration function parameters evaluated (1/SCr versus time slope, change in the CCr, ratio of SCr, and change in serum cystatin C). Among them, a significant improvement was observed in the slope for 1/SCr versus time when contrast coefficients [−1, 1, 0] were used, suggesting a valid hypothesis that the improvement in 120 μg group was remarkable. In addition, in the post-hoc analysis, a significant improvement in the ratio of SCr was achieved in the 120 μg group as well as in the 240 μg group. The difference in cystatin C levels in the treatment period also improved in the 240 μg group. Regarding the eGFR ratio, a significant improvement was observed only in the 120 μg group.
Significant effects on blood pressure or urinary protein levels were not observed in this study, and thus the mechanisms of action of BPS are thought to be different from those of existing agents such as ACEIs and ARBs. Since the majority of patients in this study was taking ACEIs and/or ARBs, TRK-100STP may be a useful for treatment of CKD patients in combination with ACEIs or ARBs. TRK-100STP was considered to be well tolerated. Most of the AEs observed in this study were similar types of the events caused by prostacyclin and its analogue.
Several important limitations to this study are apparent: as the treatment period was comparatively short (28 weeks) for evaluating the decrease in the renal function, the enrollment to the trial was restricted to those patients whose renal function had progressively declined. In addition, no dialysis or doubling of SCr was observed during the study period so that its efficacy on the renal composite endpoint including dialysis was unclear. Further evaluation in a longer study period will therefore be necessary in future.
Based on the findings of this trial, we have further been conducting a phase IIb/III trial, CASSIOPEIR (CRF Asian Study with Oral PGI2 derivative for Evaluating Improvement of Renal function) [42]. This new trial will enroll patients who have more severe disease than those enrolled in this trial in order to ensure that a sufficient number of clinical events can be observed within a designated time period, using renal composite endpoints to evaluate treatment outcome. This trial will also be dose-finding as it utilizes the same two doses as used in this study. The trial is being conducted in seven Asian countries, including Japan.

Conclusions

Although this randomized, placebo-controlled, double-blind, comparative trial failed to meet the primary endpoint to determine the recommended dose for the treatment of patients with CKD with primary glomerular disease or nephrosclerosis, it shows for the first time, that TRK-100STP might potentially prevent the decline in renal function of such patients, independent of blood pressure or urinary protein levels. These findings have helped in the design and conduct of the ongoing phase IIb/III trial, CASSIOPEIR.

Acknowledgements

This trial was sponsored by Toray Industries, Inc. and Astellas Pharma Inc. in the design, and in the collection and analysis of the data.
We would like to thank the doctors, nurses and staff at the sites listed below for their participation and assistance in the trial. We also thank Osamu Hotta and Takayuki Fujita for their important suggestion about the design of the protocol and thank Yoshitaka Mori, Takeshi Matsugi and all other members of Toray and Astellas for the participation in the trial. We would like to thank Midori Ogasawara and Satoshi Henmi of Toray, Hirotoshi Kakuta, Tomihisa Kawasaki and Hiroyuki Okumura of Astellas and Jun Kishi of MEDCORE ASSOCIATES Inc., for having provided their assistance in the preparation of the report.

Participating centers

Aizawa Hospital, Akebono Hospital, Anjo Kosei Hospital, Asahi Chuo Hospital, Chiba Seikatsu-shukanbyo Internal Medicine Clinic, Chubu Rosai Hospital, Dainohara Medical Clinic, Fujisawa City Hospital, Fujita Health University Hospital, Fukuoka University Hospital, Hamamatsu University Hospital, Hiroshima City Hospital, Hiroshima Red Cross Hospital & Atomic-bomb Survivors Hospital, Hitachi General Hospital, Hitachi Mito General Hospital, Hokkaido University Hospital, Ichikawa East Hospital, Japanese Red Cross Society Suwa Hospital, Jichi Medical University Hospital, Kasugai Municipal Hospital, Kawahara Clinic, Kawasaki Medical School Hospital, Kawashima Clinic, Kawashima Hospital, Kinashi Obayashi Hospital, Kinki Central Hospital of Mutual Aid Association of Public School Teachers, Kitasato Institute Hospital, Kitasato University Kitasato Institute Medical Center Hospital, Kobe City Medical Center General Hospital, Kyoto University Hospital, Medical Corporation Fukuseikai Fukuseikai Hospital, Mito Saiseikai General Hospital, Mitsubishi Kyoto Hospital, Munakata Medical Association Hospital, Nagasaki University Hospital of Medicine and Dentistry, Nagoya University Hospital, Nara Medical University Hospital, Narita Memorial Hospital, National Center for Global Health and Medicine, National Hospital Organization Disaster Medical Center, National Hospital Organization Kanazawa Medical Center, National Hospital Organization Kure Medical Center, National Hospital Organization Nagasaki Medical Center, National Hospital Organization Oita Medical Center, National Hospital Organization Osaka National Hospital, National Hospital Organization Shizuoka Medical Center, National Kyushu Medical Center, Nihon University Itabashi Hospital, Niigata Rinko Hospital, Niigata University Medical & Dental Hospital, NTT Medical Center Tokyo, Ohno Memorial Hospital, Okabe Hospital, Okayama Medical Center, Okayama University Hospital, Okazaki City Hospital, Osaka General Medical Center, Osaka Red Cross Hospital, Osaka University Hospital, Rinku General Medical Center, Saiki kidney clinic, Saiseikai Niigata Daini Hospital, Saiseikai Yokohamasi Nanbu Hospital, Saitama Medical University Hospital, Sakai Rumi Clinic, Sanjou General Hospital, Seirei Hamamatsu General Hospital, Sendai Shakaihoken Hospital, Shimazu Hospital, Shinrakuen Hospital, Shinshu University Hospital, Shonan Kamakura General Hospital, Showa University Fujigaoka Hospital, Social Insurance Chuo General Hospital, St. Marianna University School of Medicine Hospital, Sunagawa City Medical Center, Takayama Hospital, Tanaka Kitanoda Hospital, The University of Tokyo Hospital, Toho University Omori Medical Center, Tokai University Hospital, Tokuyama Central Hospital, Tokyo Kyosai Hospital, Tokyo Medical University Hospital, Tokyo Medical University Ibaraki Medical Center, Tosei General Hospital, Toyota Memorial Hospital, Tsuchiya General Hospital, Tsukuba Central Hospital, Tsukuba University Hospital, Ureshino Medical Center, Yokohama City Minato Red Cross Hospital, Yokohama City University Medical Center, Yokohama Minami Kyousai Hospital, Yonago Medical Center. (Alphabetical order).
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

AK and FG have no conflict of interest. TF and HO serve as a consultant of Toray. TF is a recipient of grants for Astellas, Toray, Boehringer Ingelheim, Chugai, Fukuda Denshi, Kyowa Hakko Kirin, Mitsubishi Tanabe, Mochida, Omron, Pfizer and Takeda. Also, TF is a recipient of lecture fee for Astellas, Boehringer Ingelheim, Kyowa Hakko Kirin, Mitsubishi Tanabe, Daiichi Sankyo, Novartis, Pfizer and Takeda. MI, HAKU and KO are employees of Toray, while HIKA, TK and SY are employees of Astellas.

Authors’ contributions

AK, TF and FG were assigned as medical advisors for the study but their terms of duty were all different during the study period. They were involved in the conception and design of the study, and advisers for conducting the study. HO was involved in the conception and study design and provided statistical support of the study. MI, HAKU, KO, HIKA, TK and SY were involved in the study conception and acquisition of data and data analysis. AK and HAKU wrote the draft of the manuscript. All authors critically reviewed the manuscript and approved the final manuscript as submitted.
Literatur
1.
Zurück zum Zitat Xie Y, Chen X. Epidemiology, major outcomes, risk factors, prevention and -management of chronic kidney disease in China. Am J Nephrol. 2008;28:1–7.CrossRefPubMed Xie Y, Chen X. Epidemiology, major outcomes, risk factors, prevention and -management of chronic kidney disease in China. Am J Nephrol. 2008;28:1–7.CrossRefPubMed
2.
Zurück zum Zitat Akizawa T. Current Status of Dialysis Therapy and Related Clinical Guidelines in Japan. JMAJ. 2010;53:185–7. Akizawa T. Current Status of Dialysis Therapy and Related Clinical Guidelines in Japan. JMAJ. 2010;53:185–7.
3.
Zurück zum Zitat Hwang SJ, Tsai JC, Chen HC. Epidemiology, impact and preventive care of chronic kidney disease in Taiwan. Nephrology (Carlton). 2010;15 Suppl 2:3–9.CrossRef Hwang SJ, Tsai JC, Chen HC. Epidemiology, impact and preventive care of chronic kidney disease in Taiwan. Nephrology (Carlton). 2010;15 Suppl 2:3–9.CrossRef
5.
Zurück zum Zitat KDIGO. KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. Kid Int. 2013;3:1–150. KDIGO. KDIGO 2012 clinical practice guideline for the evaluation and management of chronic kidney disease. Kid Int. 2013;3:1–150.
6.
Zurück zum Zitat ONTARGET Investigators, Yusuf S, Teo KK, Pogue J, Dyal L, Copland I, et al. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med. 2008;358:1547–59.CrossRef ONTARGET Investigators, Yusuf S, Teo KK, Pogue J, Dyal L, Copland I, et al. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med. 2008;358:1547–59.CrossRef
7.
Zurück zum Zitat Imai E, Chan JC, Ito S, Yamasaki T, Kobayashi F, Haneda M, et al. Effects of olmesartan on renal and cardiovascular outcomes in type 2 diabetes with overt nephropathy: a multicentre, randomised, placebo-controlled study. Diabetologia. 2011;54:2978–86.CrossRefPubMedPubMedCentral Imai E, Chan JC, Ito S, Yamasaki T, Kobayashi F, Haneda M, et al. Effects of olmesartan on renal and cardiovascular outcomes in type 2 diabetes with overt nephropathy: a multicentre, randomised, placebo-controlled study. Diabetologia. 2011;54:2978–86.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Fried LF, Emanuele N, Zhang JH, Brophy M, Conner TA, Duckworth W, et al. Combined angiotensin inhibition for the treatment of diabetic nephropathy. N Engl J Med. 2013;369:1892–903.CrossRefPubMed Fried LF, Emanuele N, Zhang JH, Brophy M, Conner TA, Duckworth W, et al. Combined angiotensin inhibition for the treatment of diabetic nephropathy. N Engl J Med. 2013;369:1892–903.CrossRefPubMed
9.
Zurück zum Zitat Parving HH, Brenner BM, McMurray JJ, de Zeeuw D, Haffner SM, Solomon SD, et al. Cardiorenal end points in a trial of aliskiren for type 2 diabetes. N Engl J Med. 2012;367:2204–13.CrossRefPubMed Parving HH, Brenner BM, McMurray JJ, de Zeeuw D, Haffner SM, Solomon SD, et al. Cardiorenal end points in a trial of aliskiren for type 2 diabetes. N Engl J Med. 2012;367:2204–13.CrossRefPubMed
10.
Zurück zum Zitat Moncada S, Gryglewski R, Bunting S, Vane JR. An enzyme isolated from arteries transforms prostaglandin endoperoxidases to an unstable substance that inhibits platelet aggregation. Nature. 1976;263:663–5.CrossRefPubMed Moncada S, Gryglewski R, Bunting S, Vane JR. An enzyme isolated from arteries transforms prostaglandin endoperoxidases to an unstable substance that inhibits platelet aggregation. Nature. 1976;263:663–5.CrossRefPubMed
11.
Zurück zum Zitat Yokoyama C, Yabuki T, Shimonishi M, Wada M, Hatae T, Ohkawara S, et al. Prostacyclin-deficient mice develop ischemic renal disorders, including nephrosclerosis and renal infarction. Circulation. 2002;106:2397–403.CrossRefPubMed Yokoyama C, Yabuki T, Shimonishi M, Wada M, Hatae T, Ohkawara S, et al. Prostacyclin-deficient mice develop ischemic renal disorders, including nephrosclerosis and renal infarction. Circulation. 2002;106:2397–403.CrossRefPubMed
12.
Zurück zum Zitat Spargias K, Adreanides E, Demerouti E, Gkouziouta A, Manginas A, Pavlides G, et al. Iloprost prevents contrast-induced nephropathy in patients with renal dysfunction undergoing coronary angiography or intervention. Circulation. 2009;120:1793–9.CrossRefPubMed Spargias K, Adreanides E, Demerouti E, Gkouziouta A, Manginas A, Pavlides G, et al. Iloprost prevents contrast-induced nephropathy in patients with renal dysfunction undergoing coronary angiography or intervention. Circulation. 2009;120:1793–9.CrossRefPubMed
13.
Zurück zum Zitat Ohno K, Nagase H, Matsumoto K, Nishiyama H, Nishio S. Stereoselective synthesis of 5,6,7-trinor-4,8-inter-m-phenylene-PGI2 derivatives and their inhibitory activities to human platelet aggregation. Adv Prostaglandin Thromboxane Leukot Res. 1985;15:279–81.PubMed Ohno K, Nagase H, Matsumoto K, Nishiyama H, Nishio S. Stereoselective synthesis of 5,6,7-trinor-4,8-inter-m-phenylene-PGI2 derivatives and their inhibitory activities to human platelet aggregation. Adv Prostaglandin Thromboxane Leukot Res. 1985;15:279–81.PubMed
14.
Zurück zum Zitat Kurumatani H. Pharmacological profiles and clinical effects of oral sustained release beraprost sodium (Careload LA tablets 60 μg, Berasus LA tablets 60 μg). Nihon Yakurigaku Zasshi. 2009;133:101–11.CrossRefPubMed Kurumatani H. Pharmacological profiles and clinical effects of oral sustained release beraprost sodium (Careload LA tablets 60 μg, Berasus LA tablets 60 μg). Nihon Yakurigaku Zasshi. 2009;133:101–11.CrossRefPubMed
15.
Zurück zum Zitat Nagaya N. Orally active prostacyclin analogue for cardiovascular disease. Int Angiol. 2010;29(2 Suppl):14–8.PubMed Nagaya N. Orally active prostacyclin analogue for cardiovascular disease. Int Angiol. 2010;29(2 Suppl):14–8.PubMed
16.
Zurück zum Zitat Fine LG, Bandyopadhay D, Norman JT. Is there a common mechanism for the progression of different types of renal diseases other than proteinuria? Towards the unifying theme of chronic hypoxia. Kidney Int Suppl. 2000;75:S22–6.CrossRefPubMed Fine LG, Bandyopadhay D, Norman JT. Is there a common mechanism for the progression of different types of renal diseases other than proteinuria? Towards the unifying theme of chronic hypoxia. Kidney Int Suppl. 2000;75:S22–6.CrossRefPubMed
17.
Zurück zum Zitat Eckardt KU, Bernhardt WM, Weidemann A, Warnecke C, Rosenberger C, Wiesener MS, et al. Role of hypoxia in the pathogenesis of renal disease. Kidney Int Suppl. 2005;99:S46–51.CrossRefPubMed Eckardt KU, Bernhardt WM, Weidemann A, Warnecke C, Rosenberger C, Wiesener MS, et al. Role of hypoxia in the pathogenesis of renal disease. Kidney Int Suppl. 2005;99:S46–51.CrossRefPubMed
18.
Zurück zum Zitat Kang DH, Kanellis J, Hugo C, Truong L, Anderson S, Kerjaschki D, et al. Role of the microvascular endothelium in progressive renal disease. J Am Soc Nephrol. 2002;13:806–16.CrossRefPubMed Kang DH, Kanellis J, Hugo C, Truong L, Anderson S, Kerjaschki D, et al. Role of the microvascular endothelium in progressive renal disease. J Am Soc Nephrol. 2002;13:806–16.CrossRefPubMed
19.
Zurück zum Zitat Nangaku M. Chronic hypoxia and tubulointerstitial injury: a final common pathway to end-stage renal failure. J Am Soc Nephrol. 2006;17:17–25.CrossRefPubMed Nangaku M. Chronic hypoxia and tubulointerstitial injury: a final common pathway to end-stage renal failure. J Am Soc Nephrol. 2006;17:17–25.CrossRefPubMed
20.
Zurück zum Zitat Inoue T, Kozawa E, Okada H, Inukai K, Watanabe S, Kikuta T, et al. Noninvasive evaluation of kidney hypoxia and fibrosis using magnetic resonance imaging. J Am Soc Nephrol. 2011;22:1429–34.CrossRefPubMedPubMedCentral Inoue T, Kozawa E, Okada H, Inukai K, Watanabe S, Kikuta T, et al. Noninvasive evaluation of kidney hypoxia and fibrosis using magnetic resonance imaging. J Am Soc Nephrol. 2011;22:1429–34.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Utsunomiya Y, Ogura M, Kawamura T, Mitarai T, Maruyama N, Sakai O. Attenuation of immune complex nephritis in NZB/WF1 mice by a prostacyclin analogue. Clin Exp Immunol. 1995;99:454–60.CrossRefPubMedPubMedCentral Utsunomiya Y, Ogura M, Kawamura T, Mitarai T, Maruyama N, Sakai O. Attenuation of immune complex nephritis in NZB/WF1 mice by a prostacyclin analogue. Clin Exp Immunol. 1995;99:454–60.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Kushiro M, Shikata K, Sugimoto H, Shikata Y, Miyatake N, Wada J, et al. Therapeutic effects of prostacyclin analog on crescentic glomerulonephritis of rat. Kidney Int. 1998;53:1314–20.CrossRefPubMed Kushiro M, Shikata K, Sugimoto H, Shikata Y, Miyatake N, Wada J, et al. Therapeutic effects of prostacyclin analog on crescentic glomerulonephritis of rat. Kidney Int. 1998;53:1314–20.CrossRefPubMed
23.
Zurück zum Zitat Yamada M, Sasaki R, Sato N, Suzuki M, Tamura M, Matsushita T, et al. Amelioration by beraprost sodium, a prostacyclin analogue, of established renal dysfunction in rat glomerulonephritis model. Eur J Pharmacol. 2002;449:167–76.CrossRefPubMed Yamada M, Sasaki R, Sato N, Suzuki M, Tamura M, Matsushita T, et al. Amelioration by beraprost sodium, a prostacyclin analogue, of established renal dysfunction in rat glomerulonephritis model. Eur J Pharmacol. 2002;449:167–76.CrossRefPubMed
24.
Zurück zum Zitat Yamaguchi S, Inada C, Tamura M, Sato N, Yamada M, Itaba S, et al. Beraprost sodium improves survival rates in anti-glomerular basement membrane glomerulonephritis and 5/6 nephrectomized chronic kidney disease rats. Eur J Pharmacol. 2013;714:325–31.CrossRefPubMed Yamaguchi S, Inada C, Tamura M, Sato N, Yamada M, Itaba S, et al. Beraprost sodium improves survival rates in anti-glomerular basement membrane glomerulonephritis and 5/6 nephrectomized chronic kidney disease rats. Eur J Pharmacol. 2013;714:325–31.CrossRefPubMed
25.
Zurück zum Zitat Yamaguchi S, Goto Y, Sato N, Kurumatani H, Miymoto M, Yamada M. Beraprost sodium suppresses progression of chronic renal failure by protecting renal capillary endothelial cells and improving renal hypoxia in glomerulonephritis rats. J Am Soc Nephrol. 2013;24:868A.CrossRef Yamaguchi S, Goto Y, Sato N, Kurumatani H, Miymoto M, Yamada M. Beraprost sodium suppresses progression of chronic renal failure by protecting renal capillary endothelial cells and improving renal hypoxia in glomerulonephritis rats. J Am Soc Nephrol. 2013;24:868A.CrossRef
26.
Zurück zum Zitat Goto Y, Yamaguchi S, Tamura M, Mochizuki H, Kurumatani H, Okano K, et al. A prostacyclin analog prevents the regression of renal microvascular network by inhibiting mitochondria-dependent apoptosis in the kidney of rat progressive glomerulonephritis. Prostaglandins Other Lipid Mediat. 2014;112:16–26.CrossRefPubMed Goto Y, Yamaguchi S, Tamura M, Mochizuki H, Kurumatani H, Okano K, et al. A prostacyclin analog prevents the regression of renal microvascular network by inhibiting mitochondria-dependent apoptosis in the kidney of rat progressive glomerulonephritis. Prostaglandins Other Lipid Mediat. 2014;112:16–26.CrossRefPubMed
27.
Zurück zum Zitat Mishima K, Baba A, Matsuo M, Itoh Y, Oishi R. Protective effect of cyclic AMP against cisplatin-induced nephrotoxicity. Free Radic Biol Med. 2006;40:1564–77.CrossRefPubMed Mishima K, Baba A, Matsuo M, Itoh Y, Oishi R. Protective effect of cyclic AMP against cisplatin-induced nephrotoxicity. Free Radic Biol Med. 2006;40:1564–77.CrossRefPubMed
28.
Zurück zum Zitat Itoh Y, Yano T, Sendo T, Oishi R. Clinical and experimental evidence forprevention of acute renal failure induced by radiographic contrast media. J Pharmacol Sci. 2005;97:473–88.CrossRefPubMed Itoh Y, Yano T, Sendo T, Oishi R. Clinical and experimental evidence forprevention of acute renal failure induced by radiographic contrast media. J Pharmacol Sci. 2005;97:473–88.CrossRefPubMed
29.
Zurück zum Zitat Sakai A, Yajima M, Nishio S. Cytoprotective effect of TRK-100, a prostacyclin analogue, against chemical injuries in cultured human vascular endothelial cells. Life Sci. 1990;47:711–9.CrossRefPubMed Sakai A, Yajima M, Nishio S. Cytoprotective effect of TRK-100, a prostacyclin analogue, against chemical injuries in cultured human vascular endothelial cells. Life Sci. 1990;47:711–9.CrossRefPubMed
30.
Zurück zum Zitat Tamura M, Kurumatani H, Matsushita T. Comparative effects of beraprost, a stable analogue of prostacyclin, with PGE(1), nitroglycerin and nifedipine on canine model of vasoconstrictive pulmonary hypertension. Prostaglandins Leukot Essent Fatty Acids. 2001;64:197–202.CrossRefPubMed Tamura M, Kurumatani H, Matsushita T. Comparative effects of beraprost, a stable analogue of prostacyclin, with PGE(1), nitroglycerin and nifedipine on canine model of vasoconstrictive pulmonary hypertension. Prostaglandins Leukot Essent Fatty Acids. 2001;64:197–202.CrossRefPubMed
31.
Zurück zum Zitat Nishio S, Matsuura H, Kanai N. The in vitro and ex vivo antiplatelet effect of TRK-100, a stable prostacyclin analog, in several species. Jpn J Pharmacol. 1988;47:1–10.CrossRefPubMed Nishio S, Matsuura H, Kanai N. The in vitro and ex vivo antiplatelet effect of TRK-100, a stable prostacyclin analog, in several species. Jpn J Pharmacol. 1988;47:1–10.CrossRefPubMed
32.
Zurück zum Zitat Fujita T, Fuke Y, Satomura A, Hidaka M, Ohsawa I, Endo M, et al. PGI2 analogue mitigates the progression rate of renal dysfunction improving renal blood flow without glomerular hyperfiltration in patients with chronic renal insufficiency. Prostaglandins Leukot Essent Fatty Acids. 2001;65:223–7.CrossRefPubMed Fujita T, Fuke Y, Satomura A, Hidaka M, Ohsawa I, Endo M, et al. PGI2 analogue mitigates the progression rate of renal dysfunction improving renal blood flow without glomerular hyperfiltration in patients with chronic renal insufficiency. Prostaglandins Leukot Essent Fatty Acids. 2001;65:223–7.CrossRefPubMed
33.
Zurück zum Zitat Ishida A, Hotta O, Takuma T. Clinical efficacy of PGI2 derivative (BPS) in patients with progressive renal disease. Jpn J Nephrol. 2003;45:276. Ishida A, Hotta O, Takuma T. Clinical efficacy of PGI2 derivative (BPS) in patients with progressive renal disease. Jpn J Nephrol. 2003;45:276.
34.
Zurück zum Zitat Owada A, Suda S, Hata T. Effect of long-term administration of prostaglandin I2 in incipient diabetic nephropathy. Nephron. 2002;92:788–96.CrossRefPubMed Owada A, Suda S, Hata T. Effect of long-term administration of prostaglandin I2 in incipient diabetic nephropathy. Nephron. 2002;92:788–96.CrossRefPubMed
35.
Zurück zum Zitat Wang LN, Tang Z, Shou I, Fukui M, Tomino Y. Effects of the PGI2 analog beraprost sodium on glomerular prostanoid synthesis in rats with streptozotocin-induced diabetes. Nephron. 1996;73:637–43.PubMed Wang LN, Tang Z, Shou I, Fukui M, Tomino Y. Effects of the PGI2 analog beraprost sodium on glomerular prostanoid synthesis in rats with streptozotocin-induced diabetes. Nephron. 1996;73:637–43.PubMed
36.
Zurück zum Zitat Watanabe M, Nakashima H, Mochizuki S, Abe Y, Ishimura A, Ito K, et al. Amelioration of diabetic nephropathy in OLETF rats by prostaglandin I2 analog, beraprost sodium. Am J Nephrol. 2009;30:1–11.CrossRefPubMed Watanabe M, Nakashima H, Mochizuki S, Abe Y, Ishimura A, Ito K, et al. Amelioration of diabetic nephropathy in OLETF rats by prostaglandin I2 analog, beraprost sodium. Am J Nephrol. 2009;30:1–11.CrossRefPubMed
37.
Zurück zum Zitat Sato N, Kaneko M, Tamura M, Kurumatani H. The prostacyclin analog beraprost sodium ameliorates characteristics of metabolic syndrome in obese Zucker (fatty) rats. Diabetes. 2010;59:1092–100.CrossRefPubMedPubMedCentral Sato N, Kaneko M, Tamura M, Kurumatani H. The prostacyclin analog beraprost sodium ameliorates characteristics of metabolic syndrome in obese Zucker (fatty) rats. Diabetes. 2010;59:1092–100.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Mitch WE, Walser M, Buffington GA, Lemann Jr J. A simple method of estimating progression of chronic renal failure. Lancet. 1976;2(7999):1326–8.CrossRefPubMed Mitch WE, Walser M, Buffington GA, Lemann Jr J. A simple method of estimating progression of chronic renal failure. Lancet. 1976;2(7999):1326–8.CrossRefPubMed
39.
Zurück zum Zitat Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, et al. Effects of losartanon renal and cardiovascular outcomes in patients with type 2 diabetes andnephropathy. N Engl J Med. 2001;345:861–9.CrossRefPubMed Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, et al. Effects of losartanon renal and cardiovascular outcomes in patients with type 2 diabetes andnephropathy. N Engl J Med. 2001;345:861–9.CrossRefPubMed
40.
Zurück zum Zitat Koide K, Koshikawa S, Yamane Y. Clinical evaluation of AST-120 on suppression of progression of chronic renal failure. Multi-center, double-blind study in comparison with placebo. Clin Eval. 1987;15:527–64. Koide K, Koshikawa S, Yamane Y. Clinical evaluation of AST-120 on suppression of progression of chronic renal failure. Multi-center, double-blind study in comparison with placebo. Clin Eval. 1987;15:527–64.
41.
Zurück zum Zitat Matsuo S, Imai E, Horio M, Yasuda Y, Tomita K, Nitta K, et al. Collaborators developing the Japanese equation forestimated GFR. Revised equations for estimated GFR from serum creatinine in Japan. Am J Kidney Dis. 2009;53:982–92.CrossRefPubMed Matsuo S, Imai E, Horio M, Yasuda Y, Tomita K, Nitta K, et al. Collaborators developing the Japanese equation forestimated GFR. Revised equations for estimated GFR from serum creatinine in Japan. Am J Kidney Dis. 2009;53:982–92.CrossRefPubMed
42.
Zurück zum Zitat Nakamoto H, Fujita T, Origasa H, Isono M, Kurumatani H, Okada K, et al. A multinational phase IIb/III trial of beraprost sodium, an orally active prostacyclin analogue, in patients with primary glomerular disease or nephrosclerosis (CASSIOPEIR trial), rationale and study design. BMC Nephrol. 2014;15:153.CrossRefPubMedPubMedCentral Nakamoto H, Fujita T, Origasa H, Isono M, Kurumatani H, Okada K, et al. A multinational phase IIb/III trial of beraprost sodium, an orally active prostacyclin analogue, in patients with primary glomerular disease or nephrosclerosis (CASSIOPEIR trial), rationale and study design. BMC Nephrol. 2014;15:153.CrossRefPubMedPubMedCentral
Metadaten
Titel
Orally active prostacyclin analogue beraprost sodium in patients with chronic kidney disease: a randomized, double-blind, placebo-controlled, phase II dose finding trial
verfasst von
Akio Koyama
Toshiro Fujita
Fumitake Gejyo
Hideki Origasa
Masanao Isono
Hajimu Kurumatani
Kiyonobu Okada
Hiroyuki Kanoh
Takashi Kiriyama
Shunsuke Yamada
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
BMC Nephrology / Ausgabe 1/2015
Elektronische ISSN: 1471-2369
DOI
https://doi.org/10.1186/s12882-015-0130-5

Weitere Artikel der Ausgabe 1/2015

BMC Nephrology 1/2015 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Battle of Experts: Sport vs. Spritze bei Adipositas und Typ-2-Diabetes

11.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Im Battle of Experts traten zwei Experten auf dem Diabeteskongress gegeneinander an: Die eine vertrat die Auffassung „Sport statt Spritze“ bei Adipositas und Typ-2-Diabetes, der andere forderte „Spritze statt Sport!“ Am Ende waren sie sich aber einig: Die Kombination aus beidem erzielt die besten Ergebnisse.

Vorsicht, erhöhte Blutungsgefahr nach PCI!

10.05.2024 Koronare Herzerkrankung Nachrichten

Nach PCI besteht ein erhöhtes Blutungsrisiko, wenn die Behandelten eine verminderte linksventrikuläre Ejektionsfraktion aufweisen. Das Risiko ist umso höher, je stärker die Pumpfunktion eingeschränkt ist.

Triglyzeridsenker schützt nicht nur Hochrisikopatienten

10.05.2024 Hypercholesterinämie Nachrichten

Patienten mit Arteriosklerose-bedingten kardiovaskulären Erkrankungen, die trotz Statineinnahme zu hohe Triglyzeridspiegel haben, profitieren von einer Behandlung mit Icosapent-Ethyl, und zwar unabhängig vom individuellen Risikoprofil.

Update Innere Medizin

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