Aims
SNF472 is a calcification inhibitor that is being studied as a novel treatment for calciphylaxis and cardiovascular calcification (CVC). A first study showed acceptable safety and tolerability in a single ascending dose administration in healthy volunteers and a single dose administration in haemodialysis (HD) patients. This study aimed to assess the safety, tolerability, and pharmacokinetics/pharmacodynamics relationship of intravenous SNF472 in HD patients in a multiple ascending dose administration trial with 5 doses tested for 1 week (3 administrations) and 1 dose tested for 4 weeks (12 administrations).
Methods
This double blind, randomized, placebo‐controlled Phase 1b study investigated the safety, tolerability, pharmacokinetics and pharmacodynamics of SNF472 after repeated administrations to HD patients for up to 28 days. A pharmacodynamic assessment was performed to evaluate the potential for SNF472 to inhibit hydroxyapatite (HAP) formation. Patients were grouped into 2 cohorts, receiving multiple ascending doses for 1 week (1 to 20 mg/kg, Cohort 1) and 1 dose of 10 mg/kg for 4 weeks (Cohort 2) of intravenous SNF472.
Results
Physical status, body weight, cardiorespiratory function, body temperature and laboratory parameters were in the normal range. No clinically relevant effects on heart rate or blood pressure were observed. No abnormal electrocardiogram or QTcB period were reported. The peak plasma concentration (7.6, 16.1, 46.0 and 66.9 μg/mL for 3, 5, 12.5 and 20 mg/kg, respectively) was observed at the end of the 4‐hour infusion and thereafter concentrations declined rapidly with half‐life between 32 and 65 min. SNF472 at 10 mg/kg inhibited dose dependently HAP crystallization in plasma samples after 28 days of treatment (78% inhibition, P < .001).
Conclusions
SNF472 is safe and well tolerated in HD patients after 2 schemes: multiple ascending doses for 1 week and after repeated dosing of 10 mg/kg for 4 weeks. In both schemes, SNF472 inhibits the induction of HAP crystallization. These results provide support for the use of SNF472 as a novel treatment for CVC in end‐stage renal disease.
What is already known about this subject
Survival in haemodialysis (HD) patients is correlated with cardiovascular calcification, but no drug for the treatment of cardiovascular calcification has been approved to date. SNF472 is a new calcification inhibitor with reported efficacy in preclinical models. A recent study showed acceptable safety and tolerability in healthy volunteers and after single administration in HD patients.
What this study adds
This is a double‐blind, randomised, placebo‐controlled study to assess the safety, pharmacokinetics and pharmacodynamics of intravenous SNF472 with repeated administrations to HD patients for up to 28 days. Our results support the safety and tolerability of SNF472 at repeated dosing for 4 weeks and show potential pharmacokinetic‐dependent SNF472 efficacy against calcification, thus providing a strong rationale for further clinical development.
1. INTRODUCTION
Cardiovascular calcification (CVC) is a significant risk factor for morbidity and mortality in patients with end‐stage renal disease (ESRD) on maintenance dialysis.1 Approximately 50% of patients with chronic kidney disease die from cardiovascular events.2 A correlation between the degree and progression of CVC and cardiovascular events (including mortality) has been demonstrated.3, 4, 5, 6 Key risk factors for the development of CVC in patients on haemodialysis (HD) include hyperphosphataemia, hypercalcaemia, hyperparathyroidism and abnormal vitamin D metabolism. An imbalance between inhibitors and enhancers of CVC are central to the basic pathophysiology of this condition.7, 8 Two of the major families of drugs used in HD patients each address 1 of the many factors that can lead to vascular calcification. Phosphate binders (such as sevelamer) address the hyperphosphataemia,9 whereas calcimimetics (i.e. cinacalcet) address the hyperparathyroidism10 and consequently the hypercalcaemia. However, none of these treatments target the common final pathway in the development of CVC.
SNF472 is a selective calcification inhibitor being investigated as a new approach for the treatment of CVC in HD patients with ESRD and for the treatment of calciphylaxis. It is an intravenous (IV) formulation of myo‐inositol hexaphosphate (IP6). In vitro11 and in vivo12, 13 results show that SNF472 or IP6 prevent the formation of hydroxyapatite (HAP) crystals. SNF472 directly inhibits calcium crystal formation by binding to the surface of the crystal, and IV SNF472 administration to HD patients could mitigate the impact of the imbalance between CVC inhibitors and enhancers present in these patients. By inhibiting the final common pathway of CVC, the efficacy of SNF472 is likely to be independent of the aetiology of CVC and would be observed irrespective of plasma calcium or phosphate levels.14, 15
The safety, tolerability and pharmacokinetics (PK) of IV SNF472 was evaluated in healthy volunteers (HV) following administration of single IV doses ranging from 0.5 to 12.5 mg/kg and in HD patients after treatment with 1 simple dose.16 In that Phase I first‐time‐in‐human clinical study, the PK profile showed a Cmax proportionality and a relatively short half‐life. SNF472 was well tolerated with the exception of mild‐to‐moderate local infusion site irritation in HV.
The study reported in this paper was a Phase 1b, double‐blind, placebo‐controlled, randomized, multiple ascending dose clinical trial to evaluate the safety, tolerability and PK/pharmacodynamics (PD) of IV SNF472 in HD patients.
2. METHODS
This study was conducted at a single site in accordance with the International Conference for Harmonization E6 Guidelines for Good Clinical Practice and the Declaration of Helsinki. This study is registered at Eudra CT (Identifier 2014–005076‐29) and was approved by an Independent Ethics Committee. Written informed consent was obtained from all patients prior to the start of the study.
2.1. Patients
Patients were male or nonpregnant female stable HD patients, aged 18–80 years, inclusive. At screening, patients should have been undergoing standard HD 3 times a week for at least 6 months. Causes of exclusion included: alkaline phosphatase ≥240 IU/L; hospitalization within the previous 3 months for myocardial ischaemia, unstable angina, heart failure, peripheral vascular disease, stroke or seizures; a schedule date for a renal transplant; positive testing for hepatitis B or human immunodeficiency virus; history of drug abuse in the past 2 years or participation in another trial within 1 month prior to the first dose of study drug.
2.2. Study design
The study had 2 subject cohorts with 8 patients per cohort (Figure 1 ). Cohort 1 received multiple ascending doses of study drug for 1 week, and Cohort 2 received a fixed repeated dose for 4 weeks.
Figure 1.

Flowchart of distribution of patients in Cohort 1 A, and Cohort 2 B
Cohort 1 consisted of 10 patients (8 were planned, but 2 additional patients were recruited to replace 2 patients that discontinued the study) and involved placebo and 5 different doses of study drug (Table S1 and Figure 1). Subjects participated in 5 treatment periods (TP) of 1 week each. For the 3 highest dose groups (TP1, TP2 and TP3), subjects were randomized in each TP in a 6:2 ratio to receive SNF472 (6 patients) or placebo (2 patients), ensuring that subjects who had received placebo in a specific TP received active treatment in the other 2 TPs. Therefore, a total of 6 different placebos were studied in Cohort 1. For the 2 lowest dose groups, the 8 patients from Cohort 1 were divided into 2 additional TPs (TP4 and TP5) of 4 patients each, after a washout of at least 1 month, with no additional placebos. The Dose Escalation Safety Committee (DESC) reviewed PK, tolerability, and safety data after each week of treatment in Cohort 1 (TP1 and TP2) to decide whether to proceed with dose escalation.
In cohort 2, subjects participated in a multiple dose study. They were randomized in a 6:2 ratio to receive SNF472 (6 patients) or placebo (2 patients). Patients received a single IV dose of either SNF472 or placebo on days 1, 3, 5, 8, 10, 12, 15, 17, 19, 22, 24 and 26 during the patient's dialysis session.
2.3. Treatments
The doses for Cohort 1 were 1, 3, 5, 12.5 and 20 mg/kg. For Cohort 2, the dose was 10 mg/kg. Study drug administration consisted of an IV dose of either SNF472 or placebo infused over a 4‐hour period in the morning during the subject's dialysis session. Subjects were administered study drug 3 times per week. The appropriate volume of the study drug was administered using a 100 mL bag of saline connected to an infusion pump, which was connected directly to the dialysis machine (Cordiax 5008; Fresenius Medical Care, USA) via an IV giving set (Sendal, Spain) and an accessory heparin line (Gambro Dasco, Italy; Figure S2).
2.4. Pharmacokinetic assessments
Serial blood samples for PK were obtained at specific time points (predose and up to 8 hours postdose) during the TP. Blood samples were collected into K3EDTA anticoagulant tubes and were centrifuged at 1900 g for 10 minutes at 4°C. Plasma was stored at −80°C until IP6 quantification by UPLC‐MS using the validated method described by Tur et al.17
The PK parameters calculated were: area under the curve from 0 to the time of the last quantifiable concentration (AUC0➔t); AUC from 0 to infinity (AUCinf); percentage of AUC extrapolated from AUC0➔t (%AUCex); total body clearance (CL); maximum SNF472 concentration measured in plasma (Cmax); terminal elimination half‐life (t1/2); time at which the Cmax is measured (Tmax); volume of distribution at terminal phase (Vz); and apparent first order terminal elimination rate constant (λz).
2.5. Pharmacodynamic assessments
The PD of SNF472 administered by IV infusion was evaluated by measuring HAP crystallization.11 For this purpose, 2 samples from each subject were used for PD analysis: 1 sample obtained at baseline before SNF472 infusion and 1 sample at Tmax, obtained at the end of the 4 hours of infusion. Plasma samples were mixed in 0.15 M NaCl (pH 7.4), with 1.5 mM phosphate and 12.5 mM calcium in 96‐well plates, and the crystallization of HAP was measured spectrophotometrically at 550 nm for 30 minutes.
2.6. Safety assessments
Safety parameters included: treatment‐emergent adverse events (TEAEs); clinical laboratory and haematology; clinical chemistry (including plasma electrolytes); vital signs (pulse, respiratory rate, supine blood pressure and axillary body temperature); 12‐lead electrocardiogram parameters (including cardiac intervals, PR, QRS, QT and QTc); and physical examination. Ionized calcium was measured at Tmax in all patients to monitor patients for hypocalcaemia.
2.7. Statistical analyses
Data were collected in PostgreSQL by OpenClinica™. All statistical analyses and data manipulation were performed using SAS version 9.3 (SAS Institute Inc., Cary, NC, USA).
The PK analysis was performed using the PK population defined as all patients who received at least 1 dose of SNF472 and for whom either of the primary PK parameters (Cmax or AUC0‐t) could be calculated for at least 1 TP and for whom no major protocol deviation occurred. PK parameters of SNF472 were calculated by noncompartmental analysis methods from the concentration–time data. PK parameters were summarized descriptively using arithmetic mean, standard deviation, coefficient of variation (CV%), median, minimum, maximum, geometric mean, geometric CV% and number of observations.
To assess PD, inhibition of calcification induction was calculated as the percentage change in the slope for the linear interval between predose and postdose samples, using the following formula:
For Cohort 1, the statistical significance of the data was initially assessed by a 2‐way analysis of variance (ANOVA) considering the factors time (days) and dose (0, 1, 3, 5, 12.5 and 20 mg/kg). A significant effect of dose but not of time was obtained. Therefore, a 1‐way ANOVA was performed combining days 1 and 5 and considering only SNF472 dose as an independent variable. A posthoc analysis was applied to determine the differences between the groups involved. For Cohort 2, the statistical significance of the time‐course data was assessed by a 2‐way ANOVA considering the factors time (days) and treatment (placebo vs SNF472 10 mg/kg). A significant effect of treatment but not of time was obtained. Therefore, a Student t test was performed combining days 1, 8, 15 and 26, and considering only SNF472 treatment as a factor.
3. RESULTS
3.1. Patients and treatment
In Cohort 1, 6 patients completed all periods, 2 were prematurely withdrawn after completing TP1, leading to 2 replacement patients in TP2. In Cohort 2, 8 patients were included and all patients completed the study.
Demographic characteristics were similar for Cohort 1 and Cohort 2, with mean ages of 61.8 and 62.1 years, respectively, and mean BMI values of 28.6 and 27.5 kg/m2, respectively (Table 1 ). The majority of patients were Caucasian–non‐Hispanic (70.0% in Cohort 1 and 62.5% Cohort 2) and male (60% in Cohort 1 and 87.5% in Cohort 2). A list of concomitant medication related to chronic kidney disease‐mineral bone disorder management is shown in Table S3.
Table 1.
Demographic characteristics and alcohol consumption habits
| Variable | Statistic | Cohort 1 (n = 10) | Cohort 2 (n = 8) |
|---|---|---|---|
| Sex | |||
| n | 10 | 8 | |
| Female | 4 (40.0%) | 1 (12.5%) | |
| Male | 6 (60.0%) | 7 (87.5%) | |
| Age (y) | n | 10 | 8 |
| Mean | 61.8 | 62.1 | |
| Standard deviation | 15.1 | 10.1 | |
| Median | 65.5 | 66.0 | |
| Height (cm) | n | 10 | 8 |
| Mean | 161.8 | 169.6 | |
| Standard deviation | 5.9 | 10.3 | |
| Median | 161.0 | 172.0 | |
| Weight (kg) | n | 10 | 8 |
| Mean | 75.0 | 79.0 | |
| Standard deviation | 23.3 | 13.8 | |
| Median | 68.2 | 77.4 | |
| Body mass index (kg/m2) | n | 10 | 8 |
| Mean | 28.6 | 27.5 | |
| Standard deviation | 8.2 | 4.4 | |
| Median | 27.1 | 28.7 | |
| Race | n | 10 | 8 |
| Caucasian– Hispanic | 1 (10.0%) | 3 (37.5%) | |
| Caucasian–non‐Hispanic | 7 (70.0%) | 5 (62.5%) | |
| Other | 2 (20.0%) | 0 | |
| Alcohol consumption | n | 10 | 8 |
| Yes | 1 (10.0%) | 0 | |
| No | 9 (90.0%) | 8 (100.0%) | |
3.2. PK
In cohort 1, Cmax was observed at the end of the 4‐hour infusion (Tmax; Figure 2A and Table 2) and concentrations declined rapidly falling below the lower limit of quantitation (LLOQ; 0.5 μg/mL) by 6 hours (5 mg/kg) and 8 hours (12.5, 20 mg/kg) postdose. At a dose of 1 mg/kg, only 1 subject had measurable plasma concentration.
Figure 2.

Pharmacokinetic profile of intravenous SNF472 in haemodialysis patients in a multiple ascending dose. A, Plasma SNF472 levels at Day 1. B, SNF472 plasma Cmax at Day 1 and Day 5. Results represent mean ± standard deviation
Table 2.
Summary of pharmacokinetic parameters
| Cohort 1 | |||||
|---|---|---|---|---|---|
| Parameter | SNF472 1 mg/kg Mean (CV%) | SNF472 3 mg/kg Mean (CV%) | SNF472 5 mg/kg Mean (CV%) | SNF472 12.5 mg/kg Mean (CV%) | SNF472 20 mg/kg Mean (CV%) |
| Day 1 | |||||
| Cmax (μg/mL) | ‐ | 7.6 (66) | 16.1 (33) | 46.0 (34) | 66.9 (43) |
| Tmax (min) | ‐ | 236 (0) | 217 (22) | 200 (31) | 244 (8) |
| AUC0‐t (μg h/mL) | ‐ | 20.7 (72) | 48.7 (25) | 146.5 (37) | 282.5 (49) |
| AUC0‐∞ (μg h/mL) | ‐ | 1.4 (70) | 51.8 (26) | 158.0 (39) | 350.2 (48) |
| t1/2 (min) | ‐ | 34 (11) | 32 (26) | 52 (39) | 65 (61) |
| Vz (mL) | ‐ | 6536 (22) | 6165 (46) | 7973 (62) | 5989 (29) |
| Vss (mL) | ‐ | 11746 (34) | 9395 (33) | 8766 (65) | 7295 (13) |
| CL (mL/h) | ‐ | 16957 (86) | 8759 (35) | 6309 (32) | 4662 (48) |
| Day 5 | |||||
| Cmax (μg/mL) | 0.7a | 8.0 (50) | 19.3 (33) | 46.8 (46) | 56.0 (38) |
| Tmax (min) | 232a | 235 (1) | 239 (1) | 221 (23) | 214 (22) |
| AUC0‐t (μg h/mL) | 2.1a | 24.4 (61) | 57.0 (31) | 185.6 (55) | 206.2 (40) |
| AUC0‐∞ (μg h/mL) | 2.3a | 25.3 (61) | 59.9 (36) | 162.9 (46) | 199.2 (46) |
| t1/2 (min) | NC | 35 (34) | 32 (53) | 51 (32) | 55 (53) |
| Vz (mL) | NC | 6689 (33) | 5772 (53) | 7235 (33) | 9691 (80) |
| Vss (mL) | NC | 11356 (13) | 9324 (32) | 8927 (19) | 9165 (39) |
| CL (mL/h) | 33783a | 10603 (48) | 7603 (28) | 6196 (29) | 7476 (42) |
| Cohort 2 | ||
|---|---|---|
| SNF472 10 mg/kg | ||
| Parameter | Day 1 | Day 26 |
| Cmax (μg/mL) | 38.7 (54) | 41.5 (29) |
| Tmax (min) | 238 (2) | 237 (1) |
| AUC0‐t (μg h/mL) | 115.2 (54) | 104.2 (25) |
| AUC0‐∞ (μg h/mL) | 120.4 (53) | 106.1 (26) |
| t1/2 (min) | 39 (21) | 25 (15) |
| Vz (mL) | 5695 (24) | 4647 (25) |
| Vss (mL) | 9415 (30) | 8715 (28) |
| CL (mL/h) | 10717 (100) | 7945 (30) |
AUC, Area under the curve; AUC0‐t, Area Under the Curve from 0 to the time of the last quantifiable concentration; AUC0‐∞, Area Under the Curve from 0 to infinity; CL, Clearance; Cmax, Maximum SNF472 concentration measured in plasma; NC = not counted; t1/2, Terminal elimination half‐life; Tmax, Time at which the Cmax is measured; Vz, Volume of distribution at terminal phase; Vss, Volume of distribution at steady state.
Note: Tmax values correspond to the arithmetic mean of nominal times at which Cmax was reached (just after infusion period).
‐: all patients (n = 6) presented plasma levels below of limit of detection (0.5 μg/ml).
n = 1.
The mean Cmax, AUC0‐t, and AUC0‐∞ values of SNF472 increased with increasing doses of SNF472 on day 1 and day 5 of administration in a slightly more than dose‐proportional manner (Figure 2B). No significant differences were observed between the systemic exposure (Cmax and AUC) achieved on day 1, day 3 (Cmax), and day 5 of administration.
There was no apparent accumulation with multiple doses. Despite the short half‐life observed (32–65 min), steady state was not achieved during the 4‐hour infusion period. Based on a noncompartmental PK model, accumulation and volume of distribution calculations based on the distribution t1/2 are shown in Table 2.
In cohort 2, after repeated dosing of 10 mg/kg of SNF472, Cmax was also observed at the end of the 4‐hour infusion. Concentrations declined rapidly falling below the LLOQ at 5.5 or 8 hours postadministration (Table 2 ).
There was no accumulation in terms of Cmax and AUC after repeated dosing of 10 mg/kg.
3.3. PD
In Cohort 1, no significant effect between day of administration was observed, so the results from days 1 and 5 were pooled for each dose and analysed using a 1‐way ANOVA. A statistically significant inhibition of HAP crystallization (p ≤ .001) was observed at all the doses from 3 to 20 mg/kg (approximately 70–80% inhibition). An IC50 of 2.2 mg/kg (133 mg/patient) and an IC80 of 5.6 mg/kg (469 mg/patient) were calculated (Figure 3A). Figure 3B shows the relationship between plasma circulation levels of SNF472 at Cmax and the pharmacodynamic effect in all the samples with measurable SNF472 levels (< 0.75 μM). All the doses from 3 to 20 mg/kg increased SNF472 plasma concentrations to levels that were on the pharmacodynamic plateau.
Figure 3.

Intravenous administration of SNF472 inhibits hydroxyapatite crystallization in dialysis patient plasma ex vivo. A, Dose/pharmacodynamics relationship. Four parameter log dose (mg/kg) response modelling of the inhibition of induction of hydroxyapatite crystallization in plasma samples from dialysis patients administered increasing doses of SNF472 or placebo over 5 days. Solid line represents the dose–response curve calculated from the individual data points. Dashed lines represent the 90% confidence interval. B, Pharmacokinetics/pharmacodynamics relationship.
In Cohort 2 (10 mg/kg for 4 weeks), similar inhibition of HAP crystallization was observed at all time points analysed (Figure 4A). The inhibition of crystallization over the entire study period was 78% (Figure 4B).
Figure 4.

Inhibition of hydroxyapatite (HAP) crystallization following repeated dosing of 10 mg/kg SNF472 through the entire study period. A, Time‐course of crystallization inhibition in active patients over the 4‐week period. Solid line connects the means of every day. B, Global inhibition of crystallization throughout the study period. Results represent the mean ± standard deviation. Data were analysed by a Student t test. (*) indicates significant differences vs placebo, p ≤ .001
3.4. Safety
There were no deaths and no TEAEs that led to withdrawal from the study. None of the TEAEs in this study were considered related to the study drug.
In Cohort 1, a total of 3 TEAEs were reported for 2 patients in the SNF472 12.5 mg/kg dose group, and the SNF472 1 mg/kg and 20 mg/kg dose groups each reported 1 TEAE (Table 3). There were no TEAEs in the placebo group or in the SNF472 3 and 5 mg/kg dose groups. No TEAE by preferred term or system organ class was reported for more than 1 patient, and all TEAEs were mild in intensity.
Table 3.
Summary of treatment‐emergent adverse events (TEAEs) in Cohorts 1 and 2
| Cohort 1 | ||||||
|---|---|---|---|---|---|---|
| System organ class Preferred term | Placebo (n = 6) n (%) [#] | SNF472 1 mg/kg (n = 4) n (%) [#] | SNF472 3 mg/kg (n = 4) n (%) [#] | SNF472 5 mg/kg (n = 6) n (%) [#] | SNF472 12.5 mg/kg (n = 6) n (%) [#] | SNF472 20 mg/kg (n = 6) n (%) [#] |
| Any TEAE | 0 | 1 (25.0) [1] | 0 | 0 | 2 (33.3) [3] | 1 (16.7) [1] |
| Eye disorders | ||||||
| Ocular hyperaemia | 0 | 0 | 0 | 0 | 0 | 1 (16.7) [1] |
| Gastrointestinal disorders | ||||||
| Diarrhoea | 0 | 0 | 0 | 0 | 1 (16.7) [1] | 0 |
| Immune system disorders | ||||||
| Hypersensitivity | 0 | 0 | 0 | 0 | 1 (16.7) [1] | 0 |
| Nervous system disorders | ||||||
| Headache | 0 | 0 | 0 | 0 | 1 (16.7) [1] | 0 |
| Psychiatric disorders | ||||||
| Insomnia | 0 | 1 (25) [1] | 0 | 0 | 0 | 0 |
| Cohort 2 | ||
|---|---|---|
| System Organ Class Preferred term | Placebo (n = 6) n (%) [#] | SNF472 5 mg/kg (n = 6) n (%) [#] |
| Any TEAE | 1 (50) [1] | 2 (33.3) [3] |
| Gastrointestinal disorders | ||
| Diarrhoea | 0 | 1 (16.7) [1] |
| General disorders and administration site conditions | ||
| Pyrexia | 0 | 1 (16.7) [1] |
| Infections and infestations | ||
| Renal cyst infection | 0 | 1 (16.7) [1] |
| Psychiatric disorders | ||
| Transient psychosis | 1 (50) [1] | 0 |
In Cohort 2, 3 TEAEs were reported for 2 patients (33.3%) in the study treatment group and 50% (1 of the 2 patients) in the placebo group reported a single TEAE (a serious TEAE of transient psychosis). One of the 6 patients treated with SNF472 10 mg/kg had a serious TEAE (cyst infection). Both serious TEAEs were moderate in intensity.
Physical status, body weight, cardiorespiratory function and body temperature were in the normal range throughout the study, and no clinically relevant effects on heart rate or blood pressure were observed in any of the different TP or in the multiple dose study. Total and ionized calcium levels, phosphate levels and alkaline phosphatase activity were in the normal range, and no effect of SNF472 administration was evidenced (Table 4).
Table 4.
Biochemistry parameters in Cohort 1 subjects
| Cohort 1 | ||||
|---|---|---|---|---|
| Parameter | Placebo Mean (CV%) [n] | SNF472 5 mg/kg Mean (CV%) [n] | SNF472 12.5 mg/kg Mean (CV%) [n] | SNF472 20 mg/kg Mean (CV%) [n] |
| Days 1 and 5 | ||||
| Total calcium (mg/dL) | ||||
| Predialysis | 8.90 (7.51) [12] | 9.17 (4.47) [12] | 9.18 (4.38) [12] | 9.03 (6.04) [10] |
| Postdialysis | 10.0 (9.0) [11] | 11.0 (6.6) [12] | 11.4 (8.9) [12] | 12.1 (12.9) [10] |
| Ionised calcium (mM) | ||||
| Predialysis | 1.09 (8.04) | 1.09 (13.9) | 1.16 (5.41) | 1.17 (7.00) |
| Postdialysis | 1.26 (6.83) | 1.33 (3.66) | 1.32 (5.17) | 1.33 (3.85) |
| Phosphate | ||||
| Predialysis | 3.85 (1.84) [2] | 3.77 (22.6) [3] | ‐ | ‐ |
| Postdialysis | 1.85 (26.8) [2] | 1.8 (20.0) [3] | ‐ | ‐ |
| Alkaline phosphatase (IU/L) | ||||
| Predialysis | 63.5 (38.3) [12] | 91.3 (81.7) [12] | 90.8 (69.8) [12] | 93.1 (60.7) [9] |
| Postdialysis | 78.3 (56.8) [12] | 90.9 (74.5) [12] | 91.4 (64.4) [12] | 102 (51) [9] |
Five patients had QTcB periods above 500 ms. These changes were not attributable to SNF472 as the effect was seen in placebo and treated. No electrocardiogram changes in any subject were considered to be clinically significant by the Investigator or by an independent cardiology review.
4. DISCUSSION
The objectives of this study were to assess: (1) the safety and tolerability of SNF472 after repeated administration (multiple ascending and fixed repeated doses) in HD patients; (2) the PK characteristics and dialysability after repeated dosing in HD patients; and (3) the activity of different dose levels of SNF472 on the inhibition of the calcification response to an in vitro challenge.
In Cohort 1, ascending doses of placebo, 1, 3, 5, 12.5 and 20 mg/kg were evaluated. After completion of treatment in Cohort 1 and reviewing safety and tolerability, a dose of 10 mg/kg was selected by the DESC for Cohort 2 (repeated administration doses 3 times per week for 4 weeks).
In a previous Phase I study,18 the safety, tolerability and PK of IV SNF472 were evaluated following administration of single IV doses ranging from SNF472 0.5 to 12.5 mg/kg to HV and of a single dose of 9 mg/kg to HD patients. By comparing PK results from equivalent doses of SNF472 in HV and HD patients, the dialysability of the drug was assessed. The PK profile showed a Cmax proportionality and a relatively short half‐life. The total amount of SNF472 that was excreted in urine was <1% of the total administered dose. With the exception of mild to moderate local infusion site irritation, SNF472 was well tolerated; more importantly, infusion site irritation was not observed in HD patients and is not expected to be a safety concern for HD patients as SNF472 is diluted prior to entering the circulation.
The current manuscript reports the safety, PK and PD of SNF472 in HD patients after 2 different schemes of treatment, including 5 different doses administered for 1 week (three administrations) and 1 dose administered for 4 weeks (12 administrations). In the current study, the plasma peak concentrations were observed immediately after the end of the 4‐hour infusion (Tmax) and declined rapidly to below the LLOQ for all dose levels in both cohorts. Cmax and AUC parameters increased in a slightly more than dose‐proportional manner. In both Cohort 1 and Cohort 2, PK parameters showed that there was no accumulation and steady state was not achieved. Dialysability studies show SNF472 to have an insignificant clearance through the dialysis membrane.19 Thus, the absence of clearance due to dialysis, plus the PK parameters, indicate linearity in terms of Cmax and AUC and allow a good PK prediction in future clinical studies. In a previous Phase 1 study with HV, it was demonstrated that the total amount of SNF472 excreted in urine accounted for <1% of the total administered dose.16 Since the PK profiles of HV and HD patients are similar,16 the lack of renal function in HD patients is not expected to affect the metabolism of SNF472. Mass balance and quantitative whole‐body autoradiography studies are currently being performed in rats in order to ascertain the complete metabolism and excretion of the compound, but some data on the in vivo metabolism of IP6 in rodents has been already published. Following oral and intravenous administration of radiolabelled IP6 (3H or 14C) to rats and mice, inositol was the major circulating IP6 metabolite, representing >90% of total circulating radioactivity.20, 21, 22 Lower inositol phosphates appeared quickly in plasma but were not detectable beyond 45 minutes after dosing. An elevated expired 14CO2 (about 60% of the administered dose) was found following oral administration of 14C‐IP6 and 14C‐inositol to rats,20 and this can be explained by the conversion of inositol into D‐glucuronate and xylitol.23, 24, 25 Therefore, SNF472 is metabolized through sequential dephosphorilation (hydrolysis) and the inositol ring is oxidized to water and CO2, which is eliminated through expired air.
PD analyses in this study demonstrate that the IV infusion of SNF472 to HD patients at doses from 3 to 20 mg/kg significantly inhibits the induction of HAP crystallization in plasma samples by approximately 70–80% with an IC50 of 2.2 mg/kg (133 mg/patient) and an IC80 of 5.6 mg/kg (469 mg/patient). Repeated administration (3 times per week) of SNF472 at 10 mg/kg for 4 weeks does not affect the crystallization inhibitory potential of the compound. The crystallization inhibitory potential of SNF472 is comparable along all cohorts and is in line with previous PD studies evaluating the effects of SNF472 on HAP crystallization,11 and support the use of SNF472 for the treatment of CVC in HD patients. The design of this Phase 1b trial allowed us to evidence, for the first time, a PK‐PD relationship for SNF472 in HD patients.
IP6, the active ingredient of SNF472, has been shown to prevent CVC in a variety of animal models.12, 13, 26 Published studies12, 13, 26 and nonclinical studies with SNF472 have shown that parenteral IP6 prevents CVC by between 60% and 95% in animal models. As a therapeutic treatment, it could be used at systemic concentrations that would not provoke significant reductions in ionized calcium concentrations due to chelation. The target is the calcium that is deposited on the vessel walls and not the free calcium in solution. The inhibition of CVC by IP6 is accompanied by a reportedly positive effect on bone. Both animal models27 and epidemiological data28, 29, 30 suggest that IP6 could be an effective agent for treating osteoporosis by reducing bone mineral density loss. Animals with an IP6‐enhanced diet reduced the loss of bone mineral density caused by oestrogenic deficiency in ovariectomized Wistar rats.17
Safety analyses from this study support the safety and tolerability of IV SNF472 in HD patients. There were no deaths and no TEAEs that were considered related to the study drug in either patient cohort. The incidence of TEAEs did not increase with increasing doses of study drug. Electrocardiogram, QTcB intervals and ionized calcium levels were extensively evaluated throughout this study and showed no safety issues associated with treatment with the study drug in the HD patient population. Moreover, disturbance of QT prolongations are frequent findings among patients with chronic kidney disease, especially in ESRD patients on HD.31, 32, 33, 34 A negative correlation has been established between Ca, P and K and QT/QTc interval increase in dialysis.35, 36 Both the QTc interval and the QTc dispersion increases after dialysis,29 in the range of 50–60 ms. Ionized calcium levels were maintained in the normal range, and no clinically relevant effects on heart rate or blood pressure were observed in either patient cohort. It should be noted that the key determinant of serum calcium in HD patients is the calcium concentration of the dialysis fluid. These results complement those obtained in the previous Phase 1 trial in HV and HD patients, now demonstrating that the compound is safe for the dialysis population for a period of 4 weeks. Moreover, the compound does not show accumulation in plasma after 1 month of treatment, while it shows unchanged PD activity after 1 month of treatment and a linear PK/PD relationship.
The results from this study suppose a significant step forward in the development of SNF472 as a novel inhibitor of vascular calcification, by demonstrating the overall safety, tolerability, PK/PD correlation and nondialysability of IV SNF472. This study indicates a positive risk to benefit ratio after treatment for 4 weeks and supports further development of IV SNF472 for the treatment of progression of CVC in HD patients and for the treatment of calcification‐related diseases such as calciphylaxis. Preliminary assessments of the potential for SNF472 to inhibit calcification demonstrated a marked inhibition that is sustained with repeated dosing. Future clinical trials with adequate statistical power need to be conducted to assess the efficacy of IV SNF472 for the treatment/prevention of CVC in HD patients. Two Phase 2 clinical trials with SNF472 are currently ongoing, 1 trial in calciphylaxis patients measuring the effect of SNF472 on ulcer progression, pain and quality of life (NCT02790073, all patients have completed treatment); and 1 trial evaluating the effect of SNF472 treatment for 12 months on CVC progression in ESRD patients (NCT02966028; more than 200 of the planned 400 patients are receiving treatment).
COMPETING INTERESTS
J.P., P.H.J., M.D.F., A.Z.C. and C.S. are employees or have received honoraria from Laboratoris Sanifit SL and are shareholders at Laboratoris Sanifit SL.
CONTRIBUTORS
J.P., P.H.J. and C.S. designed the study; M.D.F., A.Z.C., F.M., V.T., J.M.C. and R.O. supervised the study and carried out experiments; J.P., P.H.J, M.D.F., C.S., F.M. and J.M.C. analysed and interpreted the data; all authors drafted and revised the paper and approved the final version of the manuscript.
Supporting information
Table S1: Summary of patient disposition in Cohort 1 and Cohort 2
Figure S2: Method of SNF472 Administration
Table S3: Concomitant medication of the patients in Cohort 1 and Cohort 2
ACKNOWLEDGEMENTS
The authors acknowledge the patients who participated in this clinical trial, the personnel of the Hospital Clínic in Barcelona for their execution of the clinical trial, the team of Trial Form Support (TFS) for monitoring the study, Marta Rodríguez and Juan Vicente Torres (Syntax for Science) for data management and Ryan Overcash for its assistance on drafting the manuscript.
This study was supported by a RETOS COLABORACIÓN RTC‐2014‐2460‐1 grant from the Spanish Ministry of Economy and Competitiveness. MDF (PTQ‐11‐04860), CS (PTQ‐11‐04872) and AZC (PTQ‐13‐06355) were co‐funded by the INNCORPORA‐Torres Quevedo subprogram of the Ministerio de Economía y Competitividad, Government of Spain.
Salcedo C, Joubert PH, Ferrer MD, et al. A phase 1b randomized, placebo‐controlled clinical trial with SNF472 in haemodialysis patients. Br J Clin Pharmacol. 2019;85:796–806. 10.1111/bcp.13863
The authors confirm that the PI for this paper is Francesc Maduell and that he had direct clinical responsibility for patients.
REFERENCES
- 1. Blacher J, Guerin AP, Pannier B, Marchais SJ, London GM. Arterial calcifications, arterial stiffness, and cardiovascular risk in end‐stage renal disease. Hypertension. 2001;38(4):938‐942. [DOI] [PubMed] [Google Scholar]
- 2. Sharaf El Din UA, Salem MM, Abdulazim DO. Vascular calcification: When should we interfere in chronic kidney disease patients and how? World journal of nephrology. 2016;5(5):398‐417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Russo D, Corrao S, Battaglia Y, et al. Progression of coronary artery calcification and cardiac events in patients with chronic renal disease not receiving dialysis. Kidney Int. 2011;80(1):112‐118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Shantouf RS, Budoff MJ, Ahmadi N, et al. Total and individual coronary artery calcium scores as independent predictors of mortality in hemodialysis patients. Am J Nephrol. 2010;31(5):419‐425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Coen G, Pierantozzi A, Spizzichino D, et al. Risk factors of one year increment of coronary calcifications and survival in hemodialysis patients. BMC Nephrol. 2010;11(1):10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Saran R, Li Y, Robinson B, et al. US Renal Data System 2015 Annual Data Report: epidemiology of kidney disease in the United States. Am J Kidney Dis. 2016;67(3 Suppl 1):A7‐A8. Svii, S1–305 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Grases F, Simonet BM, Vucenik I, et al. Absorption and excretion of orally administered inositol hexaphosphate (IP(6) or phytate) in humans. Biofactors. 2001;15(1):53‐61. [DOI] [PubMed] [Google Scholar]
- 8. Moe SM, Chen NX. Mechanisms of vascular calcification in chronic kidney disease. J Am Soc Nephrol. 2008;19(2):213‐216. [DOI] [PubMed] [Google Scholar]
- 9. Giachelli CM. Vascular calcification mechanisms. J Am Soc Nephrol. 2004;15(12):2959‐2964. [DOI] [PubMed] [Google Scholar]
- 10. Savica V, Santoro D, Monardo P, Mallamace A, Bellinghieri G. Sevelamer carbonate in the treatment of hyperphosphatemia in patients with chronic kidney disease on hemodialysis. Ther Clin Risk Manag. 2008;4(4):821‐826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Ferrer MD, Perez MM, Canaves MM, Buades JM, Salcedo C, Perello J. A novel pharmacodynamic assay to evaluate the effects of crystallization inhibitors on calcium phosphate crystallization in human plasma. Sci Rep. 2017;7(1):6858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Grases F, Sanchis P, Perello J, et al. Phytate (myo‐inositol hexakisphosphate) inhibits cardiovascular calcifications in rats. Front Biosci. 2006;11(1):136‐142. [DOI] [PubMed] [Google Scholar]
- 13. Grases F, Sanchis P, Perello J, et al. Effect of crystallization inhibitors on vascular calcifications induced by vitamin D: a pilot study in Sprague‐Dawley rats. Circ J. 2007;71(7):1152‐1156. [DOI] [PubMed] [Google Scholar]
- 14. Grases F, Sanchis P, Costa‐Bauza A, et al. Phytate inhibits bovine pericardium calcification in vitro. Cardiovasc Pathol. 2008;17(3):139‐145. [DOI] [PubMed] [Google Scholar]
- 15. Grases F, Costa‐Bauza A. Phytate (IP6) is a powerful agent for preventing calcifications in biological fluids: usefulness in renal lithiasis treatment. Anticancer Res. 1999;19(5A):3717‐3722. [PubMed] [Google Scholar]
- 16. Perello J, Joubert PH, Ferrer MD, Canals AZ, Sinha S, Salcedo C. First‐time‐in‐human randomized clinical trial in healthy volunteers and haemodialysis patients with SNF472, a novel inhibitor of vascular calcification. Br J Clin Pharmacol. 2018;84(12):2867‐2876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Tur F, Tur E, Lentheric I, et al. Validation of an LC‐MS bioanalytical method for quantification of phytate levels in rat, dog and human plasma. J Chromatogr B Analyt Technol Biomed Life Sci. 2013;928:146‐154. [DOI] [PubMed] [Google Scholar]
- 18. Perelló J, Joubert PH, Ferrer MD, Canals AZ, Sinha S, Salcedo C. First‐time‐in‐human randomized clinical trial in healthy volunteers and haemodialysis patients with SNF472, a novel inhibitor of vascular calcification. Br J Clin Pharmacol. 2018;84(12):2867‐2876. 10.1111/bcp.13752 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Perello J, Gomez M, Ferrer MD, et al. SNF472, a novel inhibitor of vascular calcification, could be administered during hemodialysis to attain potentially therapeutic phytate levels. J Nephrol. 2018;31(2):287‐296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Nahapetian A, Young VR. Metabolism of 14C‐phytate in rats: effect of low and high dietary calcium intakes. J Nutr. 1980;110(7):1458‐1472. 1980/07/01 [DOI] [PubMed] [Google Scholar]
- 21. Sakamoto K, Vucenik I, Shamsuddin AM. [3H]phytic acid (inositol hexaphosphate) is absorbed and distributed to various tissues in rats. J Nutr. 1993;123(4):713‐720. [DOI] [PubMed] [Google Scholar]
- 22. Eiseman J, Lan J, Guo J, Joseph E, Vucenik I. Pharmacokinetics and tissue distribution of inositol hexaphosphate in C.B17 SCID mice bearing human breast cancer xenografts. Metabolism. 2011;60(10):1465‐1474. [DOI] [PubMed] [Google Scholar]
- 23. Burns JJ, Trousof N, Evans C, Papadopoulos N, Agranoff BW. Conversion of myo‐inositol to D‐glucuronic acid and L‐gulonic acid in the rat. Biochim Biophys Acta. 1959;33(1):215‐219. [DOI] [PubMed] [Google Scholar]
- 24. Richardson KE, Axelrod B. Conversion of inositol to CO2 by rat‐kidney preparations. Biochim Biophys Acta. 1959;32(1):265‐267. [DOI] [PubMed] [Google Scholar]
- 25. Douglas JF, King CG. The metabolism of uniformly labeled d‐glucuronic acid‐C14 in the guinea pig. J Biol Chem. 1952;198(1):187‐194. [PubMed] [Google Scholar]
- 26. Van den Berg CJ, Hill LF, Stanbury SW. Inositol phosphates and phytic acid as inhibitors of biological calcification in the rat. Clin Sci. 1972;43(3):377‐383. [DOI] [PubMed] [Google Scholar]
- 27. Grases F, Sanchis P, Prieto RM, Perello J, Lopez‐Gonzalez AA. Effect of tetracalcium dimagnesium phytate on bone characteristics in ovariectomized rats. J Med Food. 2010;13(6):1301‐1306. [DOI] [PubMed] [Google Scholar]
- 28. Lopez‐Gonzalez AA, Grases F, Roca P, Mari B, Vicente‐Herrero MT, Costa‐Bauza A. Phytate (myo‐inositol hexaphosphate) and risk factors for osteoporosis. J Med Food. 2008;11(4):747‐752. [DOI] [PubMed] [Google Scholar]
- 29. Lopez‐Gonzalez AA, Grases F, Perello J, et al. Phytate levels and bone parameters: a retrospective pilot clinical trial. Front Biosci (Elite Ed). 2010;2:1093‐1098. [DOI] [PubMed] [Google Scholar]
- 30. Lopez‐Gonzalez AA, Grases F, Monroy N, et al. Protective effect of myo‐inositol hexaphosphate (phytate) on bone mass loss in postmenopausal women. Eur J Nutr. 2013;52(2):717‐726. [DOI] [PubMed] [Google Scholar]
- 31. Khosoosi Niaki MR, Saravi M, Oliaee F, et al. Changes in QT interval before and after hemodialysis. Caspian J Intern Med. 2013;4(1):590‐594. [PMC free article] [PubMed] [Google Scholar]
- 32. Alabd MA, El‐Hammady W, Shawky A, Nammas W, El‐Tayeb M. QT Interval and QT Dispersion in Patients Undergoing Hemodialysis: Revisiting the Old Theory. Nephron extra. 2011;1(1):1‐8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Di Iorio B, Molony D, Bell C, et al. Sevelamer versus calcium carbonate in incident hemodialysis patients: results of an open‐label 24‐month randomized clinical trial. Am J Kidney Dis. 2013;62(4):771‐778. [DOI] [PubMed] [Google Scholar]
- 34. Nie Y, Zou J, Liang Y, et al. Electrocardiographic abnormalities and QTc interval in patients undergoing hemodialysis. PloS one. 2016;11(5):e0155445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Genovesi S, Vincenti A, Rossi E, et al. Atrial fibrillation and morbidity and mortality in a cohort of long‐term hemodialysis patients. Am J Kidney Dis. 2008;51(2):255‐262. [DOI] [PubMed] [Google Scholar]
- 36. Di Iorio B, Torraca S, Piscopo C, et al. Dialysate bath and QTc interval in patients on chronic maintenance hemodialysis: pilot study of single dialysis effects. J Nephrol. 2012;25(5):653‐660. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Table S1: Summary of patient disposition in Cohort 1 and Cohort 2
Figure S2: Method of SNF472 Administration
Table S3: Concomitant medication of the patients in Cohort 1 and Cohort 2
