Next Article in Journal
Phytochemical Characterization and Assessment of the Wound Healing Properties of Three Eurasian Propolis
Previous Article in Journal
A New LC-MS/MS-Based Method for the Simultaneous Detection of α-Tocopherol and Its Long-Chain Metabolites in Plasma Samples Using Stable Isotope Dilution Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

A Phase 1 Study to Assess the Pharmacokinetics, Food Effect, Safety, and Tolerability of Sepiapterin in Healthy Japanese and Non-Japanese Participants

PTC Therapeutics, Warren, NJ 07059, USA
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2024, 17(11), 1411; https://doi.org/10.3390/ph17111411
Submission received: 26 August 2024 / Revised: 12 October 2024 / Accepted: 19 October 2024 / Published: 22 October 2024
(This article belongs to the Section Pharmacology)

Abstract

:
Background: Sepiapterin is a natural precursor of tetrahydrobiopterin (BH4), a key cofactor for phenylalanine hydroxylase. It is being developed for the treatment of patients with phenylketonuria. In this study, the ethnic differences in pharmacokinetics and safety of sepiapterin in Japanese and non-Japanese participants and food effects were evaluated. Methods: Healthy participants (n = 60) received a single oral dose of sepiapterin at either 20, 40, or 60 mg/kg with a low-fat diet. The Japanese participants received two doses at 40 mg/kg, either under fasted conditions or with a low-fat diet with a 3-day washout period in between. Results: Sepiapterin was well tolerated in all participants, with no serious adverse events. Sepiapterin was quickly absorbed (Tmax 1.4–4.5 h) and rapidly and extensively converted to BH4 (Tmax ~4 h). Exposures to sepiapterin were <1% of BH4. BH4 exposures were essentially dose-independent between 20 and 60 mg/kg. A low-fat diet increased BH4 exposures in Japanese participants by 1.7-fold compared with fasted conditions. Conclusions: BH4 exposures (Cmax and AUC0–last) in Japanese participants were 10–30% higher than in non-Japanese participants, which is deemed not clinically relevant; no dose adjustment is warranted. The slightly higher BH4 exposures in Japanese participants are likely due to the higher frequency of ABCG2 c.421C>A mutation in the Japanese population.

1. Introduction

Phenylketonuria (PKU) is a rare inborn error of metabolism disease caused by reduced functionality of phenylalanine hydroxylase (PAH), an essential enzyme converting phenylalanine (Phe) to tyrosine [1,2]. PAH with its cofactor tetrahydrobiopterin (BH4) metabolizes Phe to tyrosine. In PKU, where PAH is deficient, elevated blood Phe levels and corresponding increased Phe in the brain can result in neurocognitive deficits [3]. PKU is diagnosed at birth with the near-universal adoption of newborn screening. Currently, there is no cure for PKU. The initial management of PKU consists of a prompt institution of a stringent dietary Phe restriction supplemented with specifically designed low-Phe medical formula (amino acid powder) and foods. However, dietary management alone faces great challenges in adherence, especially in older children, adolescents, and adults where the Phe-restricted diet causes nutritional inadequacy and social isolation.
Sepiapterin is a novel oral small molecule currently in clinical development for the treatment of pediatric and adult patients with PKU. Sepiapterin is a natural precursor of BH4, and its administration enhances BH4 concentrations. Sepiapterin and BH4 both bind to PAH to correct and stabilize the conformational structure of PAH, hence enhancing the activity of variant PAH. The binding of either BH4 or sepiapterin and the effects on the PAH enzyme are PAH variant-specific [4,5]. Sepiapterin has been shown to efficiently reduce blood Phe in Phase 2 and Phase 3 studies in patients with PKU [6,7]. In the recently completed Phase 3 APHENITY study (NCT05099640), children and adult participants with PKU were treated with orally administered sepiapterin at 60 mg/kg/day in a fed state and showed clinically significant benefits [7]. Of 156 participants, 114 (73%) responded to sepiapterin treatment (the response was defined as a reduction of ≥15% blood Phe level from baseline). A significant reduction of 64.2% from the baseline blood Phe concentration was observed among 98 participants in the primary analysis set, which included all participants 2 years and older who achieved ≥30% reduction in blood Phe following the 14-day sepiapterin responsiveness test during Part 1 of the study.
The pharmacokinetics (PK) of sepiapterin and its metabolite BH4 were previously examined in adult healthy volunteers in a first-in-human study following single and multiple ascending oral doses of sepiapterin and in a relative bioavailability and food effects study [2,8]. Oral sepiapterin is quickly absorbed and rapidly and extensively converted to BH4. The sepiapterin maximum-observed plasma concentration (Cmax) and area under the plasma concentration–time curve from the start of dosing to the last quantifiable concentration (AUC0–last) were generally <1% of the BH4 values in healthy volunteers. Plasma BH4 reached Cmax around 4 h (Tmax) and returned to near-baseline endogenous concentrations by 24 h. Administering sepiapterin with low-fat and high-fat diets increased BH4 AUC from the start of dosing to 24 h (AUC0–24h) by 1.7-fold and 2.8-fold, respectively. No apparent accumulation of BH4 was observed after 7-day repeated daily dosing at doses up to 60 mg/kg with a high-fat diet. BH4 exposures increased approximately dose proportionally within the dose range of 5 to 20 mg/kg and far less than dose proportionally in the dose range of 20 to 60 mg/kg when administered with a high-fat diet. The highest doses that have been studied in healthy volunteers and participants with PKU are a single dose of 80 mg/kg under fasted conditions and repeated doses of 60 mg/kg QD (once daily) with a high-fat diet [2,8].
PKU has been described in all ethnic groups, and its incidence worldwide varies widely. It was estimated that 0.45 million individuals have PKU worldwide [9]. In Japan, it occurs in approximately 1 in every 125,000 subjects [9]. To shorten the delay of introduction of innovative drugs into Japan, it is recommended in ICH E5(R1) and Japan PMDA (PSB/PED Notification No. 1225-2) to investigate potential interethnic differences by enrolling Japanese participants into multi-regional clinical trials (MRCTs).
In all previous clinical studies of sepiapterin, both in healthy volunteers and patients with PKU, the majority of participants were white; only a very limited number were Asian. The aim of this Phase 1 study was to evaluate potential differences in the PK and safety profile of sepiapterin in Japanese participants compared to non-Japanese participants. The PK profile, food effect, and safety and tolerability of three doses of sepiapterin (20, 40, and 60 mg/kg) in Japanese and non-Japanese healthy participants were evaluated. These doses were selected as these were the doses being investigated in the Phase 3 studies in patients with PKU 2 years and older.
Administering sepiapterin with food increases exposure to sepiapterin and BH4 [8]. Hence, PKU patients have generally been instructed to take sepiapterin with food during treatment. The fat composition of the Phe-restricted diet for PKU patients is similar to that of a low-fat diet. Hence, during this study, sepiapterin was administered with a low-fat meal in all cohorts. In the cohort with the 40 mg/kg dose, all Japanese participants, received an additional dose under the fasted condition to further explore the food effect in the Japanese population.

2. Results

2.1. Demographic Characteristics

A total of 60 participants enrolled and completed the study. Errors in blood sample collection were identified after completion of Cohorts 1 and 2, and therefore, additional participants were enrolled in Cohorts 4 and 5 as replacements. All 60 participants were included in the safety analysis set, and only participants (n = 36) in Cohorts 3, 4, and 5 were included in the PK analysis set. Of the thirty-six participants (eighteen Japanese and eighteen non-Japanese) enrolled in Cohorts 3, 4, and 5 for PK assessment, two participants (Japanese) were excluded from the PK analysis for the fed condition due to deviation from the low-fat diet on the day of dosing.
The mean age of participants was 32.0 years (range 20–55 years), the mean body weight was 66.4 kg (range 47.6–90.1 kg), and the mean BMI was 22.9 kg/m2 (range 18.5–30.0 kg/m2) (Table 1). Comparable numbers of male (n = 34) and female (n = 26) participants were enrolled. Baseline demographics were generally similar between Japanese and non-Japanese participants.

2.2. Pharmacokinetics

Predose endogenous sepiapterin concentrations in plasma were below the lower limit of quantitation (LLOQ, 0.75 ng/mL), while the mean endogenous BH4 concentrations in plasma were in the range of 2.01 to 2.95 ng/mL (Table 2). Small fluctuations of endogenous plasma BH4 concentration were observed, with slightly higher concentrations in the afternoon compared to in the morning (Table 2). The endogenous BH4 in Japanese participants (2.39 and 2.95 ng/mL in the morning and afternoon, respectively) was slightly higher than in non-Japanese participants (2.01 and 2.50 ng/mL in the morning and afternoon, respectively). Given that the baseline level of endogenous BH4 was considered relatively negligible compared to BH4 Cmax following sepiapterin administration (<5%), the PK parameters of BH4 without baseline correction are presented here.
Sepiapterin (20 to 60 mg/kg dose range) was quickly absorbed following oral administration with Cmax (2.1 to 4.9 ng/mL) reached between 1.0 and 5.0 h (Tmax) (Table 3). Absorbed sepiapterin was rapidly and extensively converted to BH4, and the sepiapterin concentrations in plasma dropped below the LLOQ by 12 h for the majority of participants (Figure 1). Sepiapterin exposures (Cmax and AUC0–last) were generally <1% of its metabolite BH4 (Table 3). Due to the rapid elimination, sepiapterin T1/2 and AUC0–inf could not be reliably estimated.
BH4 Cmax in plasma was reached between 4 and 5 h (Tmax) (Table 3), and concentrations declined steadily following the Tmax. In some individuals, this decline was biphasic, with the second phase starting approximately between 12 and 24 h post-dose (Figure 1). As a result, the terminal T1/2 for BH4 could not be reliably estimated for some participants due to the insufficient number of time points available in the terminal phase. For participants with estimable terminal T1/2, mean values ranged between 6 and 7.6 h (Table 3).

2.2.1. Ethnic Comparison

There was no apparent difference in the absorption rate between Japanese and non-Japanese participants, and the Tmax for BH4 ranged between 3.0 and 5.0 h for both populations (Table 3).
BH4 exposure was slightly higher in Japanese participants compared with non-Japanese participants; GMRs (%) of BH4 exposures (Cmax, AUC0–last, and AUC0–inf) were between 110% and 129% (Table 4). This difference is not considered clinically relevant, and therefore, no dose adjustment is required for Japanese participants.

2.2.2. Food Effects

In Japanese participants, the administration of sepiapterin with food increased BH4 exposures relative to fasted conditions (Table 5). BH4 AUC0–last and Cmax were approximately 1.7-fold higher with a low-fat diet compared with fasted conditions (Figure 2).

2.2.3. Dose Proportionality

No differences in dose proportionality were observed between Japanese and non-Japanese participants. In the dose range of 20 to 60 mg/kg of sepiapterin, negligible increases in BH4 exposure were observed with an increased dose. Using the power model, the slope (90% CI) for natural log-transformed BH4 AUC0–inf, AUC0–last, and Cmax was 0.026 (−0.094, 0.145), 0.064 (−0.042, 0.170), and 0.017 (−0.101, 0.134), respectively (Figure 2). The 90% CI for all three parameters contained zero. Similarly, negligible increases in sepiapterin exposure were observed with increased sepiapterin dose.

2.3. Safety

No deaths or serious AEs occurred during this study, and no participant withdrew due to a TEAE. The incidence of TEAEs was comparable between Japanese and non-Japanese participants, and there was no relationship with sepiapterin dose (Table 6). A total of 18 participants (30%) experienced 33 TEAEs. Of the thirty-three TEAEs, twenty-eight were mild (Grade 1) and five were moderate (Grade 2) in severity. Headache was the most commonly reported TEAE (four (13.3%) Japanese participants and three (10.0%) non-Japanese participants). Of the TEAEs, 14 were considered probably or possibly related to sepiapterin. There were no clinically relevant changes in laboratory evaluations, vital signs, and 12-lead ECG measurements.

3. Discussion

Exposure to BH4 was slightly higher in Japanese participants than in non-Japanese participants, and sepiapterin and BH4 PK parameters were generally comparable with previous studies in which the majority of participants were white [2,8]. Sepiapterin was rapidly absorbed, quickly and extensively converted to BH4, and declined to below the LLOQ generally by 12 h postdose. The Cmax and AUC0–last of sepiapterin were generally <1% of those of BH4, with BH4 having a terminal T1/2 of approximately 6 h in most participants.
In this study, BH4 exposure was found to be independent of sepiapterin dose (20 to 60 mg/kg) in both Japanese and non-Japanese participants. This deviated from previous clinical studies conducted in the same sepiapterin dose range, in which an increase in BH4, albeit far less than dose-proportional, was observed [2,8]. However, small, less than dose-proportional increases in BH4 exposure were observed in the dose range of 20 to 40 mg/kg sepiapterin in Japanese participants in this study. For Japanese participants, BH4 Cmax and AUC0–last increased from 485 to 567 ng/mL and 2830 to 3490 h·ng/mL, respectively, representing an increase of 16.9% and 17.5% when the dose was increased from sepiapterin 20 to 40 mg/kg administered with a low-fat diet. This degree of increase in BH4 is consistent with clinical studies in which most participants were white [2,8]. In a previous study, in which 78% of participants were white, BH4 Cmax and AUC0–24h increased from 317 to 519 ng/mL and 2010 to 3140 h·ng/mL, respectively, when the dose, administered with a low-fat low-calorie meal, was increased from 20 to 60 mg/kg [8]. This represents an increase of 63.7% in BH4 Cmax and 56.2% in BH4 AUC0–24h with a 3-fold increase in sepiapterin dose. The independence of BH4 exposures from doses observed in this study, across the sepiapterin dose range from 20 to 60 mg/kg, suggests that the absorption of the drug was likely saturated at the higher end of the dose range and there was only a negligible difference in exposure without an overall central trend, considering the interindividual variability and the small sample size.
PK analysis demonstrated that BH4 exposures were slightly higher (10% to 29%) in Japanese participants compared with non-Japanese participants. However, this modest increase is deemed not clinically relevant, and a dose adjustment is not required for the Japanese population. The higher BH4 exposures in Japanese participants could potentially be attributed to a higher frequency of the ABCG2 c.421C>A polymorphism in Asian populations.
Both sepiapterin and BH4 have been shown to be substrates of the breast cancer resistance protein (BCRP), also known as the ABCG2 transporter. It is known that polymorphisms in BCRP contribute to interindividual differences in exposures to orally administered drugs that are substrates of BCRP [10,11,12,13,14]. It is hypothesized that the slightly higher BH4 exposures in Japanese participants observed in this study could be due to the higher reported frequency of the ABCG2 c.421C>A mutation present in Eastern Asian populations (34% to 45%) in comparison to white populations (11% to 17%) [15,16,17], although the BCRP genotype was not collected in this study to confirm this hypothesis. Clinical studies have demonstrated that people have increased exposures to rosuvastatin, an indicator substrate of BCRP, when having the BCRP genotype ABCG2 c.421C/C, c.421C/A, and c.421A/A, in that order. Thus, the higher exposures of rosuvastatin and some other BCRP substrates, such as sulfasalazine, fluvastatin, atorvastatin, and sunitinib [18], in Eastern Asians, such as Japanese and Chinese subjects, are attributed to allele frequency in BCRP mutations.
In a drug–drug interaction study of sepiapterin, it was found that BH4 Cmax, AUC0–last, and AUC0–inf in participants with the ABCG2 c.421C/A genotype were 1.36-fold, 1.39-fold, and 1.38-fold higher, respectively, than in participants with the c.421C/C genotype [19]. The Cmax, AUC0–last, and AUC0–inf for rosuvastatin were found to be 1.60-fold, 1.61-fold, and 1.52-fold higher, respectively, in participants with the c.421C/A genotype compared to participants with the c.421C/C genotype in the same study [19].
It is proposed that the BCRP genotype information be collected in future clinical studies of sepiapterin to explore the reasons for the difference in BH4 exposures between Japanese and non-Japanese populations, or more widely, between Eastern Asian and white populations.
If such inter-ethnic differences in BH4 exposures are confirmed to be due to the ABCG2 c.421C>A polymorphism, sepiapterin has the potential to be a better clinical indicator compound for pharmacogenomic/pharmacogenetic studies than the currently widely used rosuvastatin for the following reasons: sepiapterin is not a substrate of OATP1B1 (organic anion transporting peptide member 1B1), so it is unlikely to be impacted by OATP1B1 polymorphism as is the case with rosuvastatin; there is negligible biliary excretion of sepiapterin, as demonstrated in a 14C-labeled rat mass balance study (data on file); and sepiapterin metabolism is mainly mediated by sepiapterin reductase, dihydrofolate reductase, and aromatic amino acid hydroxylases, and therefore, it is unlikely to be impacted by polymorphisms of CYP2C9 or CYP3A4/5 [1].
Examination of the PK of BH4 in previous clinical studies has shown a monophasic decline of BH4 concentrations up to 24 h post-dose [2]. However, in this study reported here, where PK samples were collected up to 32 h post-dose, a biphasic decline in BH4 concentrations post-Tmax was observed in some participants, with the second phase starting approximately 12 to 24 h post-dose. Due to insufficient duration of blood sample collection, the T1/2 could not be reliably estimated for these participants; however, if the T1/2 of the second phase were to be calculated, the value could be as high as 49.6 h (with a span of 0.322). It is postulated that the second phase of elimination is likely related to the release of BH4 trapped deep in peripheral tissues. This appears to have a minimal impact on the overall BH4 exposures, given no apparent accumulation of BH4 has been consistently demonstrated in healthy adults and patients with PKU at repeated doses of sepiapterin up to 60 mg/kg administered with food [2,8].
Examination of the PK of BH4 in Japanese participants following administration of sepiapterin 40 mg/kg with a low-fat low-calorie meal showed that exposures (Cmax and AUC0–t) were increased by approximately 1.7-fold compared with fasted conditions. This is consistent with observations in other ethnic populations (mainly participants who were white) where administration of sepiapterin with a low-fat diet increased BH4 exposures by approximately 1.7-fold and with a high-fat diet increased BH4 exposures by approximately 2.8-fold [8].

4. Methods

4.1. Ethics

This Phase 1 study was conducted at a single clinical research facility managed by LabCorp (Leeds, UK) in full compliance with the protocol, and the principles of the International Council for Harmonization (ICH) Good Clinical Practice (GCP), the Declaration of Helsinki, and applicable Medicines and Healthcare Products Regulatory Agency (MHRA) regulatory requirements. The study protocol was approved by the Independent Ethics Committees (IEC) of Fast Track REC (Research Ethic Committee), London, UK; London-Harrow REC, Bristol, UK; and the MHRA prior to initiation. All participants provided written informed consent prior to entering the study.

4.2. Study Design

This was a Phase 1, open-label, single-dose study of sepiapterin in 60 healthy adult males and females (30 Japanese and 30 non-Japanese) equally divided into five cohorts, with each cohort composed of six Japanese and six non-Japanese participants (Figure 3). Participants received a single dose of sepiapterin at 20 mg/kg in Cohorts 1 and 4, 40 mg/kg in Cohorts 2 and 5, and 60 mg/kg in Cohort 3. Sepiapterin was dosed with a low-fat diet with compositions consistent with the US Food and Drug Administration (FDA) guidance for all participants in Cohorts 1, 3, and 4, and all non-Japanese participants in Cohorts 2 and 5 [20]. Japanese participants in Cohorts 2 and 5 were randomized into one of the two sequences, and sepiapterin (40 mg/kg) was administered either with a low-fat diet or under the fasted condition in one of the two periods with a 3-day washout period in between. Cohorts 4 and 5 were the replacements for Cohorts 1 and 2 due to errors during blood sample collection for Cohorts 1 and 2 that resulted in non-evaluable PK data.
All participants fasted overnight for at least 10 h and received sepiapterin in either a fasted condition or fed state (within 30 min after starting the breakfast (low-fat low-calorie diet) and 5 min after consumption of food). Intake of food was restricted for at least 4 h post-dose, and fluid intake was restricted from 1 h prior to dose to 2 h post-dose (except for those foods and liquids provided in the meal and provided for dose administration). Additional restrictions included no consumption of xanthine/caffeine from 24 h prior to dose to 24 h post-dose and alcohol from 48 h prior to dosing to discharge from the clinical site.

4.3. Participants

For this study, 30 healthy Japanese participants and 30 healthy non-Japanese participants were enrolled across the cohorts (Figure 3). Key inclusion criteria included the following: healthy adult male and female participants aged 18–55 years (inclusive) with a body mass index (BMI) ranging from 18.5 to 30.0 kg/m2 (inclusive). Japanese participants must have had two Japanese biological parents and four Japanese grandparents, not lived outside of Japan for more than 10 years at the time of screening, and had no significant change in diet since leaving Japan. For non-Japanese participants, it was required that they were of European, white Latin American, or African descent. Key exclusion criteria included pregnancy, lactation, or suffering from any medical condition that could confound the results of the study. Females were required to have a negative pregnancy test at screening and be either abstinent from sex or use at least 2 forms of contraception during the study.
Matching of Japanese and non-Japanese participants occurred at the cohort level on sex, cohort mean age (within ±10 years), and BMI (within ±6 kg/m2) at the screening visit.

4.4. Pharmacokinetic Sample Collection and Bioanalytical Methods

Whole blood samples (3.0 mL each) were obtained on the day prior to each sepiapterin administration (between 2:00 p.m. and 8:00 p.m. (BH4 only)), pre-dose (within 1 h before dosing), and 0.25, 0.5, 1, 2, 3, 4, 5, 6, 8, 12, 16, 24, and 32 h post-dose via venipuncture into Vacutainer® tubes prefilled with K2EDTA as anticoagulant and put on wet ice. Detailed procedures regarding the collection, storage of plasma samples, and quantitative determination of sepiapterin and BH4 in plasma using a validated HPLC-MS/MS method were described previously [21].

4.5. Data Analysis

4.5.1. Safety and Tolerability

All adverse events (AEs) were coded using the Medical Dictionary for Regulatory Activities version 24.1. All AEs were assigned a severity grade using Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0. Treatment-emergent adverse events (TEAEs) and drug-related AEs were assessed daily for the duration of the study and at the safety follow-up visit. Evaluations included clinical laboratory tests (chemistry, hematology, urinalysis), physical examination, 12-lead electrocardiograms (ECGs), and vital signs (blood pressure, respiratory rate, body temperature, and heart rate).

4.5.2. Statistical Analyses

No formal sample size calculations were performed; the number of participants (six Japanese and six non-Japanese/cohort) was chosen based on feasibility.
Non-compartmental analysis of plasma concentration–time data was performed using Phoenix® WinNonlin, version 8.1.1 or higher (Certara, Princeton, NJ, USA). AUC was calculated using the linear up logarithm–down trapezoidal method. Regression was performed on the terminal elimination phase of the individual semi-logarithm concentration-time data. At least three non-zero post-Tmax concentrations, the span ≥ 1.5-fold of the estimated T1/2, and adjusted R2 ≥ 0.80 were required for the regression. AUC0–inf was estimated only if T1/2 could be reliably determined, and the extrapolated AUC was ≤20%.
For analysis of ethnic variance between Japanese and non-Japanese participants, the Cmax, AUC0–last, and AUC0–inf of sepiapterin and BH4 were analyzed using an analysis of variance (ANOVA) model following natural log-transformation of PK parameters. The model included treatment group and race as fixed effects, whereas the participant was a random effect. For each PK parameter, the LSM (least square mean) for each group, the difference in LSMs between Japanese and non-Japanese participants, and corresponding 90% confidence intervals (CIs) were calculated. These values were back-transformed to calculate the geometric least square mean (GLSM), geometric mean ratio (GMR) of GLSMs, and corresponding 90% CI for the ratio of GLSM.
The effect of food on PK parameters (Cmax, AUC0–last, and AUC0–inf) in Japanese participants was assessed using the same mixed-effects model as described for the ethnic analysis, with feeding condition (fasted or fed) as a fixed effect instead of race.
Dose proportionality was explored using the power model that included race as a fixed effect with natural logarithm-transformed PK parameters (Cmax, AUC0–last, and AUC0–inf) [22].
ln(parameter) = intercept + slope × ln(dose) + race + error
Dose proportionality was concluded if the 90% CI for the slope of regression was within the range of 0.80 and 1.25, and dose independence was concluded if the 90% CI contained zero.

5. Conclusions

Sepiapterin was well tolerated by both Japanese and non-Japanese healthy adults following single oral doses of up to 60 mg/kg. All TEAEs were mild (twenty-eight out of thirty-three) or moderate (five out of thirty-three) in severity. BH4 exposures in Japanese participants were 10% to 29% higher than in non-Japanese participants following oral administration of sepiapterin in the dose range of 20 to 60 mg/kg with a low-fat diet; however, this is not deemed to be clinically relevant, and no dose adjustment is required for Japanese participants. The slightly higher BH4 exposures in Japanese participants are hypothesized to be due to the higher frequency of the ABCG2 c.421C>A mutation in the Japanese population. In Japanese participants, BH4 exposures were 1.7-fold higher when sepiapterin was administered at 40 mg/kg with a low-fat diet compared with fasted conditions. The increase in BH4 exposures was negligible in the sepiapterin dose range of 20 to 60 mg/kg.

Author Contributions

Conceptualization, design, and analysis, L.G., N.S. and R.K.; sample analysis, D.K.; medical monitoring, K.I.; supervision, N.S. and R.K.; first draft, L.G.; review and edit: all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by PTC Therapeutics, Inc.

Institutional Review Board Statement

This study was originally approved by the Fast Track REC (London). After the initial REC was disbanded, this study was reassigned to London-Harrow REC for protocol amendment 1. It was conducted from 26 January 2022 to 12 November 2022 at a single site (LabCorp CRU Limited, Leeds, UK). This prospective study adhered to the principles of the Declaration of Helsinki and Good Clinical Practice guidelines.

Informed Consent Statement

Informed consent was obtained from all participants involved in this study.

Data Availability Statement

The datasets generated and/or analyzed during this current study are available from the corresponding author upon reasonable request.

Acknowledgments

Medical writers (Sarah Milner and Elizabeth Goodwin, PTC Therapeutics) provided editorial assistance in the preparation of this manuscript.

Conflicts of Interest

All authors are employees of PTC Therapeutics Inc. and own stocks in the company.

References

  1. Werner, E.R.; Blau, N.; Thony, B. Tetrahydrobiopterin: Biochemistry and Pathophysiology. Biochem. J. 2011, 438, 397–414. [Google Scholar] [CrossRef] [PubMed]
  2. Smith, N.; Longo, N.; Levert, K.; Hyland, K.; Blau, N. Phase I Clinical Evaluation of CNSA-001 (Sepiapterin), A Novel Pharmacological Treatment for Phenylketonuria and Tetrahydrobiopterin Deficiencies, in Healthy Volunteers. Mol. Genet. Metab. 2019, 126, 406–412. [Google Scholar] [CrossRef] [PubMed]
  3. van Wegberg, A.M.J.; MacDonald, A.; Ahring, K.; Bélanger-Quintana, A.; Blau, N.; Bosch, A.M.; Burlina, A.; Campistol, J.; Feillet, F.; Giżewska, M.; et al. The Complete European Guidelines on Phenylketonuria: Diagnosis and Treatment. Orphanet J. Rare Dis. 2017, 12, 162. [Google Scholar] [CrossRef] [PubMed]
  4. Underhaug, J.; Aubi, O.; Martinez, A. Phenylalanine Hydroxylase Misfolding and Pharmacological Chaperones. Curr. Top. Med. Chem. 2012, 12, 2534–2545. [Google Scholar] [CrossRef]
  5. Gersting, S.; Barroso, M.; Etzold, S.; Smith, N. The Effects of the Pharmacological Chaperone Sepiapterin on Phenylalanine Hydroxylase Activity of Common Variants in Phenylketonuria. Genet. Med. Open 2023, 1, 17. [Google Scholar] [CrossRef]
  6. Bratkovic, D.; Margvelashvili, L.; Tchan, M.; Nisbet, J.; Smith, N. PTC923 (Sepiapterin) Lowers Elevated Blood Phenylalanine in Subjects with Phenylketonuria: A Phase 2 Randomized, Multi-Center, Three-Period Crossover, Open-Label, Active Controlled, All-Comers Study. Metabolism 2022, 128, 155116. [Google Scholar] [CrossRef]
  7. Muntau, A.C.; Longo, N.; Ezgu, F.; Schwartz, I.V.D.; Lah, M.; Bratkovic, D.; Margvelashvili, L.; Kiykim, E.; Zori, R.; Campistol Plana, J.; et al. Effects of oral sepiapterin on blood phenylalanine levels in a broad range of patients with phenylketonuria: Results of a phase 3, randomised, double-blind, placebocontrolled trial (APHENITY). Lancet 2024, 404, 1333–1345. [Google Scholar] [CrossRef]
  8. Gao, L.; Kaushik, D.; Xia, Y.; Ingalls, K.; Milner, S.; Smith, N.; Kong, R. Relative oral bioavailability and food effects of two sepiapterin formulations in healthy participants. Clin. Pharmacol. Drug Dev. 2024, 13, 506–516. [Google Scholar] [CrossRef]
  9. Hillert, A.; Anikster, Y.; Belanger-Quintana, A.; Burlina, A.; Burton, B.K.; Carducci, C.; Chiesa, A.E.; Christodoulou, J.; Đorđević, M.; Desviat, L.R.; et al. The Genetic Landscape and Epidemiology of Phenylketonuria. Am. J. Hum. Genet. 2020, 107, 234–250. [Google Scholar] [CrossRef]
  10. Ieiri, I. Functional Significance of Genetic Polymorphisms in P-Glycoprotein (MDR1, ABCB1) and Breast Cancer Resistance Protein (BCRP, ABCG2). Drug Metab. Pharmacokinet. 2012, 27, 85–105. [Google Scholar] [CrossRef]
  11. Birmingham, B.K.; Bujac, S.R.; Elsby, R.; Azumaya, C.T.; Wei, C.; Chen, Y.; Mosqueda-Garcia, R.; Ambrose, H.J. Impact of ABCG2 and SLCO1B1 polymorphisms on pharmacokinetics of rosuvastatin, atorvastatin and simvastatin acid in Caucasian and Asian subjects: A class effect? Eur. J. Clin. Pharmacol. 2015, 71, 341–355. [Google Scholar] [CrossRef] [PubMed]
  12. Zhang, W.; Yu, B.N.; He, Y.J.; Fan, L.; Li, Q.; Liu, Z.Q.; Wang, A.; Liu, Y.L.; Tan, Z.R.; Fen, J.; et al. Role of BCRP 421C>A polymorphism on rosuvastatin pharmacokinetics in healthy Chinese males. Clin. Chim. Acta 2006, 373, 99–103. [Google Scholar] [CrossRef] [PubMed]
  13. Keskitalo, J.E.; Zolk, O.; Fromm, M.F.; Kurkinen, K.J.; Neuvonen, P.J.; Niemi, M. ABCG2 Polymorphism Markedly Affects the Pharmacokinetics of Atorvastatin and Rosuvastatin. Clin. Pharmacol. Ther. 2009, 86, 197–203. [Google Scholar] [CrossRef] [PubMed]
  14. Wan, Z.; Wang, G.; Li, T.; Xu, B.; Pei, Q.; Peng, Y.; Sun, H.; Cheng, L.; Zeng, Y.; Yang, G.; et al. Marked Alteration of Rosuvastatin Pharmacokinetics in Healthy Chinese with ABCG2 34G>A and 421C>A Homozygote or Compound Heterozygote. J. Pharmacol. Exp. Ther. 2015, 354, 310–315. [Google Scholar] [CrossRef]
  15. Wu, H.F.; Hristeva, N.; Chang, J.; Liang, X.; Li, R.; Frassetto, L.; Benet, L.Z. Rosuvastatin Pharmacokinetics in Asian and White Subjects Wild Type for Both OATP1B1 and BCRP Under Control and Inhibited Conditions. J. Pharm. Sci. 2017, 106, 2751–2757. [Google Scholar] [CrossRef]
  16. Li, R.; Barton, H.A. Explaining Ethnic Variability of Transporter Substrate Pharmacokinetics in Healthy Asian and Caucasian Subjects with Allele Frequencies of OATP1B1 and BCRP: A Mechanistic Modeling Analysis. Clin. Pharmacokinet. 2018, 57, 491–503. [Google Scholar] [CrossRef]
  17. Chen, L.; Manautou, J.E.; Rasmussen, T.P.; Zhong, X.B. Development of precision medicine approaches based on inter-individual variability of BCRP/ABCG2. Acta Pharm. Sin. B 2019, 9, 659–674. [Google Scholar] [CrossRef]
  18. Karibe, T.; Imaoka, T.; Abe, K.; Ando, O. Curcumin as an In Vivo Selective Intestinal Breast Cancer Resistance Protein Inhibitor in Cynomolgus Monkeys. Drug Metab. Dispos. 2018, 46, 667–679. [Google Scholar] [CrossRef]
  19. Gao, L.; Kaushik, D.; Ingalls, K.; Smith, N.; Kong, R. Clinical Assessment of Breast Cancer Resistance Protein (BCRP)-Mediated Drug Drug Interactions of Sepiapterin with Curcumin and Rosuvastatin in Healthy Volunteers. Drugs R D 2024, 24, 477–487. [Google Scholar] [CrossRef]
  20. FDA. Guidance for Industry: Assessing the Effects of Food on Drugs in INDs and NDAs-Clinical Pharmacology Considerations. Available online: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/assessing-effects-food-drugs-inds-and-ndas-clinical-pharmacology-considerations (accessed on 24 August 2024).
  21. Kaushik, D.; Gao, L.; Yuan, K.; Tang, B.; Kong, R. LC-MS/MS Methods for Direct Measurement of Sepiapterin and Tetrahydrobiopterin in Human Plasma and Clinical Applications. Bioanalysis 2024, 16, 75–89. [Google Scholar] [CrossRef]
  22. Hummel, J.; McKendrick, S.; Brindley, C.; French, R. Exploratory Assessment of Dose Proportionality: Review of Current Approaches and Proposal for a Practical Criterion. Pharm. Stat. 2009, 8, 38–49. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Mean (SD) sepiapterin and BH4 concentrations following oral administration of 40 mg/kg Sepiapterin in non-Japanese and Japanese participants. BH4, tetrahydrobiopterin; SD, standard deviation. Non-Japanese participants received the dose with a standard low-fat low-calorie meal. Japanese participants received two single doses separated by a 4-day washout in between either under a fasted condition or with a low-fat low-calorie meal in each period in their specific treatment sequence.
Figure 1. Mean (SD) sepiapterin and BH4 concentrations following oral administration of 40 mg/kg Sepiapterin in non-Japanese and Japanese participants. BH4, tetrahydrobiopterin; SD, standard deviation. Non-Japanese participants received the dose with a standard low-fat low-calorie meal. Japanese participants received two single doses separated by a 4-day washout in between either under a fasted condition or with a low-fat low-calorie meal in each period in their specific treatment sequence.
Pharmaceuticals 17 01411 g001
Figure 2. Dose linearity and food effects of BH4 AUC0–last (A) and Cmax (B) following sepiapterin administration in non-Japanese and Japanese participants. AUC0–last, area under the plasma concentration–time curve from the start of dosing to the last quantifiable concentration; BH4, tetrahydrobiopterin; Cmax, maximum observed plasma concentration.
Figure 2. Dose linearity and food effects of BH4 AUC0–last (A) and Cmax (B) following sepiapterin administration in non-Japanese and Japanese participants. AUC0–last, area under the plasma concentration–time curve from the start of dosing to the last quantifiable concentration; BH4, tetrahydrobiopterin; Cmax, maximum observed plasma concentration.
Pharmaceuticals 17 01411 g002
Figure 3. Study design. Fed: low-fat low-calorie diet Cohorts 4 and 5 were replacements for Cohorts 1 and 2 due to errors during blood sample collection for Cohorts 1 and 2 that resulted in non-evaluable PK data.
Figure 3. Study design. Fed: low-fat low-calorie diet Cohorts 4 and 5 were replacements for Cohorts 1 and 2 due to errors during blood sample collection for Cohorts 1 and 2 that resulted in non-evaluable PK data.
Pharmaceuticals 17 01411 g003
Table 1. Baseline demographics of study population.
Table 1. Baseline demographics of study population.
VariablePK Population (n = 36) aOverall b
(n = 60)
Non-Japanese
(n = 18)
Japanese
(n = 18)
Age (years), Median (min, max)31.0 (20, 54)35.5 (25, 51)32.0 (20, 55)
Sex, n (%)
  Male11 (61.1)10 (55.6)34 (56.7)
  Female7 (38.9)8 (44.4)26 (43.3)
Race, n (%)
  Asian018 (100)30 (50.0)
  White15 (83.3)026 (43.3)
  Black or African American2 (11.1)03 (5.0)
  Other1 (5.6)01 (1.7)
Body weight (kg), mean (min, max)74.0 (56.1, 90.1)61.7 (47.6, 81.2)66.4 (47.6, 90.1)
BMI (kg/m2), mean (min, max)24.4 (20.4, 28.2)21.5 (18.8, 25.6)22.9 (18.5, 30.0)
BMI, body mass index; PK, pharmacokinetics. a Participants with valid concentration measurements for PK evaluation from Cohorts 3, 4, and 5. b All participants enrolled in the study from Cohorts 1, 2, 3, 4, and 5.
Table 2. Endogenous BH4 plasma concentrations in Japanese and non-Japanese participants.
Table 2. Endogenous BH4 plasma concentrations in Japanese and non-Japanese participants.
ParameterJapaneseNon-Japanese
p.m.a.m.p.m.a.m.
n17181818
Mean (SD)2.95 (0.667)2.39 (0.477)2.50 (0.428)2.01 (0.344)
CV%22.619.917.117.1
Median (min, max)2.81 (1.98, 4.30)2.37 (1.79, 3.60)2.38 (1.82, 3.31)2.01 (1.15, 2.66)
SD, standard deviation; CV%, percentage of covariance; a.m.: the morning time; p.m.: the afternoon to evening time.
Table 3. Mean (SD) of BH4 and sepiapterin PK parameters in non-Japanese and Japanese participants.
Table 3. Mean (SD) of BH4 and sepiapterin PK parameters in non-Japanese and Japanese participants.
ParameterSepiapterin (20 mg/kg)Sepiapterin (40 mg/kg)Sepiapterin (60 mg/kg)
Non-Japanese FedJapanese FedNon-Japanese FedJapanese FedJapanese FastedNon-Japanese FedJapanese Fed
(n = 6)(n = 6)(n = 6)(n = 4)d(n = 6)(n = 6)(n = 6)
BH4
AUC0–last, h·ng/mL2670 (150)2970 (607)2830 (662)3490 (772) 2200 (311)2720 (491)3290 (234)
AUC0–inf, h·ng/mL2710 (140)3100 (660) b2790 (679) b3390 (885) c2370 (500) c2580 (487) c3260 (205) d
Cmax, ng/mL420 (37)485 (109)438 (99)567 (148)340 (46)403 (81)514 (60)
Tmax a, h4.0 (4.0, 5.0)5.0 (4.0, 5.0)4.5 (3.0, 5.0)5.0 (4.1, 5.0)5.0 (4.0, 5.0)4.0 (3.0, 5.0)4.0 (3.0, 5.0)
T1/2, h6.07 (0.941)6.35 (0.781) b7.78 (2.08) b6.88 (1.33) c9.83 (2.14) c7.57 (1.59) c6.78 (1.44) d
Sepiapterin
AUC0–last, h·ng/mL8.1 (3.5)11.8 (4.3)13.3 (9)15.9 (4.8)10.3 (4.2)13.1 (6.9)25.3 (12.8)
Cmax, ng/mL2.1 (0.8)2.4 (0.8)2.7 (1.2)3.4 (0.4)2.7 (0.9)2.4 (1)4.9 (1.5)
Tmaxa, h3 (1.0, 4.0)4.5 (3.0, 5.0)3.5 (2.0, 12.0)3 (2.0, 5.0)1 (0.5, 3.0)2 (1.0, 4.0)4 (2.0, 5.0)
AUC0–inf, area under the concentration–time curve from time zero extrapolated to infinity; AUC0–last, area under the plasma concentration–time curve from the start of dosing to the last quantifiable concentration; BH4, tetrahydrobiopterin; Cmax, maximum observed plasma concentration; PK, pharmacokinetics; SD, standard deviation; T1/2, apparent terminal elimination half-life; Tmax, time to maximum observed plasma concentration. a Tmax is presented as median (minimum, maximum). b n = 5. c n = 3. d n = 4. Two participants were excluded from the analysis due to deviation from the low-fat diet on the day of dosing.
Table 4. Geometric mean ratios of BH4 exposures in Japanese and non-Japanese participants following a single oral dose of sepiapterin.
Table 4. Geometric mean ratios of BH4 exposures in Japanese and non-Japanese participants following a single oral dose of sepiapterin.
ParameterSepiapterin
Dose
BH4 GLSMBH4 GMR,
% (90% CI)
Non-Japanese
(Reference)
Japanese
(Test)
AUC0–last (h·ng/mL)20 mg/kg26632920110 (92–130)
40 mg/kg27623422124 (102–150)
60 mg/kg26853284122 (103–145)
AUC0–inf (h·ng/mL)20 mg/kg27093002111 (93–132)
40 mg/kg27243315122 (98–152)
60 mg/kg25523256128 (101–161)
Cmax (ng/mL)20 mg/kg419476114 (94–137)
40 mg/kg429552129 (104–159)
60 mg/kg396511129 (107–156)
AUC0–inf, area under the concentration–time curve from time zero extrapolated to infinity; AUC0–last, area under the plasma concentration–time curve from the start of dosing to the last quantifiable concentration; BH4, tetrahydrobiopterin; CI, confidence interval; Cmax, maximum observed plasma concentration; GLSM, geometric least square mean; GMR, geometric mean ratio.
Table 5. Effects of food on BH4 exposures in Japanese participants following a single dose of 40 mg/kg sepiapterin.
Table 5. Effects of food on BH4 exposures in Japanese participants following a single dose of 40 mg/kg sepiapterin.
ParameterBH4 GLSMBH4 GMR %
(90% CI)
Fasted (Reference) Fed a (Test)
(n = 4)(n = 4)
AUC0–last (h·ng/mL)19623290168 (122–230)
Cmax (ng/mL)320532167 (109–254)
AUC0–last, area under the plasma concentration–time curve from the start of dosing to the last quantifiable concentration; BH4, tetrahydrobiopterin; CI, confidence interval; Cmax, maximum observed plasma concentration; GLSM, geometric least square mean; GMR, geometric mean ratio. a Fed = low-fat low-calorie diet.
Table 6. Incidence of treatment-emergent adverse events.
Table 6. Incidence of treatment-emergent adverse events.
Incidence, Number of Participants (%)/Number of TEAEs
Cohorts 1 and 4 Fed Non-JapaneseCohorts 1 and 4 Fed JapaneseCohorts 2 and 5 Fed Non-JapaneseCohorts 2 and 5 Fasted JapaneseCohorts 2 and 5 Fed JapaneseCohort 3 Fed Non-JapaneseCohort 3 Fed JapaneseOverall
20 mg/kg40 mg/kg60 mg/kg
n12121212126660
Any TEAE2 (16.7)/34 (33.3)/43 (25.0)/85 (41.7)/54 (33.3)/91 (16.7)/31 (16.7)/118 (30.0)/33
  Mild2 (16.7)/24 (33.3)/43 (25.0)/65 (41.7)/54 (33.3)/81 (16.7)/21 (16.7)/118 (30.0)/28
  Moderate1 (8.3)/102 (16.7)/201 (8.3)/11 (16.7)/105 (8.3)/5
  Severe00000000
  Life-threatening00000000
TEAE related to drug1 (8.3)/13 (25.0)/33 (25.0)/74 (33.3)/42 (16.7)/41 (16.7)/11 (16.7)/114 (23.3)/21
TEAE leading to discontinuation00000000
TEAE, treatment-emergent adverse event.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Gao, L.; Kaushik, D.; Ingalls, K.; Smith, N.; Kong, R. A Phase 1 Study to Assess the Pharmacokinetics, Food Effect, Safety, and Tolerability of Sepiapterin in Healthy Japanese and Non-Japanese Participants. Pharmaceuticals 2024, 17, 1411. https://doi.org/10.3390/ph17111411

AMA Style

Gao L, Kaushik D, Ingalls K, Smith N, Kong R. A Phase 1 Study to Assess the Pharmacokinetics, Food Effect, Safety, and Tolerability of Sepiapterin in Healthy Japanese and Non-Japanese Participants. Pharmaceuticals. 2024; 17(11):1411. https://doi.org/10.3390/ph17111411

Chicago/Turabian Style

Gao, Lan, Diksha Kaushik, Kimberly Ingalls, Neil Smith, and Ronald Kong. 2024. "A Phase 1 Study to Assess the Pharmacokinetics, Food Effect, Safety, and Tolerability of Sepiapterin in Healthy Japanese and Non-Japanese Participants" Pharmaceuticals 17, no. 11: 1411. https://doi.org/10.3390/ph17111411

APA Style

Gao, L., Kaushik, D., Ingalls, K., Smith, N., & Kong, R. (2024). A Phase 1 Study to Assess the Pharmacokinetics, Food Effect, Safety, and Tolerability of Sepiapterin in Healthy Japanese and Non-Japanese Participants. Pharmaceuticals, 17(11), 1411. https://doi.org/10.3390/ph17111411

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop