A Phase IIa Clinical Trial of

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Pediatric Drugs (2020) 22:95–104

https://doi.org/10.1007/s40272-019-00373-3

ORIGINAL RESEARCH ARTICLE

A Phase IIa Clinical Trial of 2‑Iminobiotin for the Treatment of Birth


Asphyxia in DR Congo, a Low‑Income Country
Thérèse Biselele1 · Jephté Bambi1 · Dieu M. Betukumesu1 · Yoly Ndiyo1 · Gabriel Tabu2 · Josée Kapinga2 ·
Valérie Bola3 · Pascal Makaya3 · Huibert Tjabbes4 · Peter Vis5 · Cacha Peeters‑Scholte4

Published online: 21 January 2020


© Springer Nature Switzerland AG 2020

Abstract
Aim The main burden of hypoxic-ischemic encephalopathy falls in low-income countries. 2-Iminobiotin, a selective inhibi-
tor of neuronal and inducible nitric oxide synthase, has been shown to be safe and effective in preclinical studies of birth
asphyxia. Recently, safety and pharmacokinetics of 2-iminobiotin treatment on top of hypothermia has been described. Since
logistics and the standard of medical care are very different in low-resource settings, the aim of this study was to investigate
safety and pharmacokinetics of Two-IminoBiotin in the Democratic Republic of Congo (TIBC).
Methods Near-term neonates, born in Kinshasa, Democratic Republic of Congo, with a Thompson score ≥ 7 were eligible
for inclusion. Excluded were patients with (1) inability to insert an umbilical venous catheter for administration of the study
drug; (2) major congenital or chromosomal abnormalities; (3) birth weight < 1800 g; (4) clear signs of infection; and (5) mori-
bund patients. Neonates received six infusions of 2-iminobiotin 0.16 mg/kg started within 6 h after birth, with 4-h intervals,
targeting an AUC​0–4h of 365 ng*h/mL. Safety, defined as vital signs, the need for clinical intervention after administration
of study drug, occurrence of (serious) adverse events, and pharmacokinetics were assessed.
Results After parental consent, seven patients were included with a median Thompson score of 10 (range 8–16). No relevant
changes in vital signs were observed over time. There was no need for clinical intervention due to administration of study
drug. Three patients died, two after completing the study protocol, one was moribund at inclusion and should not have been
included. Pharmacokinetic data of 2-iminobiotin were best described using a two-compartment model. Median AUC​0–4h was
664 ng*h/mL (range 414–917). No safety issues attributed to the administration of 2-iminobiotin were found.
Conclusion The present dosing regimen resulted in higher AUCs than targeted, necessitating a change in the dose regimen
in future efficacy trials. No adverse effects that could be attributed to the use of 2-iminobiotin were observed.
EudraCT number 2015-003063-12.

Key Points
Electronic supplementary material The online version of this
article (https​://doi.org/10.1007/s4027​2-019-00373​-3) contains 2-Iminobiotin, a selective nitric oxide synthase inhibitor,
supplementary material, which is available to authorized users. is a potential neuroprotective drug after birth asphyxia.

* Cacha Peeters‑Scholte No safety issues were found after administration of


cacha.peeters@neurophyxia.com 2-iminobiotin for birth asphyxia in DR Congo, a low-
1
income country.
Neonatal Unit, Department of Pediatrics, University Hospital
of Kinshasa, Kinshasa, Democratic Republic of Congo The dose regimen of 2-iminobiotin should be adjusted to
2
Neonatal Unit, Department of Pediatrics, Clinique Ngaliema, meet the predefined target AUC.
Kinshasa, Democratic Republic of Congo
3
Neonatal Unit, Department of Pediatrics, Hôpital Saint
Joseph, Kinshasa, Democratic Republic of Congo
4
Neurophyxia BV, Onderwijsboulevard 225,
5223 DE ’s‑Hertogenbosch, The Netherlands
5
LAP&P Consultants, Leiden, The Netherlands

Vol.:(0123456789)
96 T. Biselele et al.

1 Introduction term neonates with moderate to severe HIE with a dose of


0.2 mg/kg/dose every 4 h [11]. Five out of the six neonates
The world has made substantial progress in reducing child experienced severe HIE. 2-IB was well tolerated, espe-
mortality in the past several decades. However, globally cially no signs of hypotension were observed and no rel-
2.5 million newborns still died in 2017, and neonatal evant changes in hematology or biochemistry were noted,
deaths accounted for 47% of all deaths of under-5-year- other than those that were related to the condition of HIE.
olds, increasing from 41% in 2000 [1]. Twenty-three per- Two patients died because of hypoxic-ischemic-related
cent of these deaths are being caused by birth asphyxia brain injury. In all patients, no (severe) adverse events
[2]. Additionally, birth asphyxia leads to a substantial that could be attributed to 2-IB were reported. Although
burden of care due to long-term neurological disability limited PK data were available, 2-IB exposure in terms of
and impairment. AUC​0–4h was higher than expected (566–712 ng*h/mL).
Currently, there are no established neuroprotective In retrospect, the estimated allometric exponent of 1.0,
treatment strategies for birth asphyxia in low-income based on interspecies scaling, was too high [10]. Using
countries (LIC). Standard care mainly consists of medi- adult human data with an accepted allometric coefficient
cation to relieve symptoms of pain, epilepsy, and infection. of 0.75 and correcting for immaturity would have been
In the past few years, therapeutic hypothermia (TH; whole more precise in determining the neonatal clearance and
body hypothermia or selective head cooling) has been thereby the dose [12]. The Data Safety Monitoring Board
introduced to reduce morbidity and mortality following (DSMB) advised to conservatively decrease the 2-IB dose
birth asphyxia in term-born neonates in the Western world. from 0.2 to 0.16 mg/kg/dose every 4 h in the current TIBC
However, in LIC, treatment with TH has not proven its study based on the sparse PK data obtained in neonates
efficacy yet [3], and a rescue treatment is urgently needed. and simulation experiments (see also supplemental Table 1
2-Iminobiotin (2-IB), a selective inhibitor of induc- in the electronic supplementary material [ESM] [13]; H.
ible and neuronal nitric oxide synthase and a vitamin B7 Tjabbes, of Neurophyxia B.V., personal communication).
analog with anti-apoptotic properties, has recently been Since logistics and the standard of medical care are very
evaluated as a potential drug to reduce the devastating different in LIC compared with the Western world, the aim
effects of hypoxic-ischemic encephalopathy (HIE) [4]. of the current TIBC study was to study the safety and PK of
After being tested in two piglet HIE models [5, 6], several 2-IB in a phase IIa clinical study in normothermic neonates
models in rats [7, 8], and in a human phase I study in after moderate to severe birth asphyxia in the Democratic
adult male volunteers [9], it was shown that 2-IB had an Republic Congo (DRC), a LIC.
excellent safety profile and was well tolerated up to high
dosages.
Based on the dose-escalation study in newborn piglets 2 Methods
subjected to HIE, and treated with a 2-IB dose of 0.1, 0.2,
or 1.0 mg/kg every 4 h or placebo, it was shown that all 2-IB 2.1 Setting
doses significantly improved short-term outcome. Short-
term outcome was defined as increased survival with a nor- This trial was performed in three large hospitals in Kinshasa,
mal amplitude integrated electroencephalography (aEEG), the capital of the DRC, between June and October 2016.
and decreased nitrotyrosine staining of parietal cortex, tem- The University Hospital of Kinshasa is in the south-western
poral cortex, and thalamus at 48 h after HIE [6]. A dose part of Kinshasa. The Clinique Ngaliema is a well-equipped
comparable to a 0.2 mg/kg dose in the piglet with a target hospital in the north-west of Kinshasa, and the Saint Joseph
area under the concentration–time curve from 0 to 4 h (AUC​ Hospital is a conventional Catholic hospital in the center of
0–4h) of 365 ng*h/mL was suggested to be the most appropri-
Kinshasa. There is no ventilation equipment available in the
ate dose for use in future human clinical HIE trials. three centers.
When combining pharmacokinetics (PK) data obtained A medical mobile team was alerted when an asphyxiated
from both animal models and adult humans, clearance baby was admitted in one of the three hospitals. Asphyxia
appeared to increase linearly with body weight and a con- was defined as a Thompson score of 7 or higher at 1–3 h
stant mg/kg dose achieved similar exposure across dif- of age. The Thompson score is a clinical measure of the
ferent species [10]. Therefore, a dose of 0.2 mg/kg every severity of the HIE and ranges between 0 (normal) and 22
4 h was predicted to be the optimal dose for a study in (severely affected) [14]. The medical mobile team helped
normothermic asphyxiated neonates. doctors on duty to obtain informed consent, and in the con-
For that reason, a pilot study was conducted to investi- duct of trial-related procedures. Written informed consent
gate the safety and pharmacokinetics of 2-IB in six (near) was obtained from one of the parents in the local language
(Lingala) or French before start of treatment. When the
2-Iminobiotin for Birth Asphyxia in Low-Income Countries 97

consenting parent was illiterate, informed consent was given the study drug; (2) major congenital or chromosomal abnor-
verbally in the presence of an impartial witness. malities such as diaphragmatic hernia, omphalocele, clini-
cally identified trisomy 13 or trisomy 18; (3) birth weight
2.2 Study Design < 1800 g; (4) clear signs of infection (defined as a rectal
temperature > 37.5 °C or skin rash such as pustules); (5)
Dual ethical review was performed in both the sponsor and moribund patients.
the host country. The protocol was approved by the ethics
committee of the University of Kinshasa, DRC and received 2.4 Study Drug
a positive advice from the ethics committee of Utrecht, the
Netherlands. The study was registered at http://www.clini​ 2-IB is a stable small molecule organic compound (243 Da)
caltr ​ialsr​egist​er.eu (EudraCT number 2015-003063-12). with an adequate aqueous solubility at lower pH values.
This study is a phase IIa clinical study investigating the There are no metabolites formed either in humans or piglets,
safety and pharmacokinetics of 2-IB administered to neo- and the compound is completely cleared via the kidney. The
nates with moderate to severe birth asphyxia treated with compound is a high clearance compound with a volume of
standard care without hypothermia in a low-resource setting. distribution slightly above total body water volume and the
An independent DSMB, consisting of three independ- kinetics appeared linear.
ent members with sufficient expertise in the clinical care 2-IB drug product formulations were temperature-logged
of neonates with perinatal asphyxia, biostatistics in early- transported to DRC and stored in a chocolate cooler (not
phase clinical studies, medical ethics, and pharmacology, exceeding temperatures of 25 °C) in the university hospi-
advised on the safety of study subjects and the overall sci- tal in Kinshasa. Earlier stability studies using storing at
entific merit of trial continuation, and assessed the results 25 ± 2 °C/60% relative humidity (RH), 30 ± 2 °C/65% RH
of this clinical phase IIa study. The strategy for safety sur- and 40 ± 2 °C/75% RH showed a very stable 2-IB drug prod-
veillance by the DSMB was defined in the DSMB charter uct. Samples were tested for appearance, pH, osmolality,
as follows: (a) The DSMB reviewed the safety data during content, impurities, (sub)visible particles, closure integrity,
the two planned meetings; also, the sponsor and the Coor- and sterility. All parameters remained unchanged during the
dinating Investigator asked the DSMB to review individual course of 6 months at 40 ± 2 °C/75% RH and of 5 years at
serious adverse events (SAEs). (b) The DSMB informed on 25 ± 2 °C/60% RH and 30 ± 2 °C/65% RH. No increase of
any suspected unexpected serious adverse reaction (SUSAR) impurities/degradation products were observed over the time
that had occurred involving 2-IB within 7 days for fatal and course of the studies (data not shown).
life-threatening SUSARs and 15 days for other SUSARs. (c)
However, at any time during the study, if there was convinc- 2.5 Dose Regimen
ing evidence of harm based on the (unblinded) data includ-
ing, but not limited to, SAEs and SUSARs, the DSMB could After parental consent, neonates received six IV infusions of
recommend that the sponsor consider such options as (but 0.16 mg/kg 2-IB, with 4-hourly intervals. 2-IB was admin-
not limited to) halting the study temporarily or definitely or istered as a slow intravenous bolus in a solution of 0.75 mg/
adapting the dosage regimen for all or selected populations. mL via an umbilical catheter. The first dose had to be admin-
(d) The DSMB made a recommendation to the sponsor after istered within 6 h after birth.
review of the data. The stopping rule for safety was defined
as discontinuation of the study because major safety issues 2.6 Outcome
were found that were related to the use of the study drug.
Additionally, the DSMB of the current PK and safety study For safety analysis, the following parameters were
also advised on the ultimate dose regimen to be used in a monitored:
future randomized controlled phase IIb study, investigating
the efficacy of 2-IB for moderate to severe birth asphyxia 1. Vital signs (heart frequency, respiratory rate, and non-
in LIC. invasive blood pressure) using a vital signs monitor
(Mindray Bio-Medical Electronics, iMEC series, Shen-
2.3 Patients zhen, China) from before start until 4 h after the last
study drug administration, at 36 h and 48 h after the first
Near-term neonates (gestational age ≥ 36 weeks) with a study drug administration.
Thompson score ≥ 7 between 1 and 3 h after birth and the 2. Need for clinical intervention due to respiratory and cir-
ability to start treatment within 6 h after birth were included culatory insufficiency from the start until 15 min after
in this study. Exclusion criteria were as follows: (1) inability administration of the study drug.
to insert an umbilical venous catheter for administration of
98 T. Biselele et al.

3. The occurrence of AEs/SAEs, including mortality. Given the small dataset, a formal statistical covariate
analysis was not considered to be reliable, hence a graphi-
2.7 Pharmacokinetic Analysis cal exploratory analysis of the influence of selected covari-
ates (birth weight, gestational age, and Thompson score)
For PK analysis, five PK samples of 0.4 mL were taken via of interest on clearance was performed for the subjects
heel prick; directly after completion of the first infusion, in the current TIBC study. Additionally, individual clear-
just before the second and last infusion, and 1 h and 4 h ance estimates from the subjects in the model develop-
after the last infusion. Blood samples were centrifuged as ment dataset were compared between the 2-STEP study
soon as possible after sampling, and the supernatant was and the current TIBC study. The main aim of the modeling
frozen at − 20 °C until temperature-logged transfer to the exercise was to obtain individual model-based exposure
Netherlands for determination of the concentrations. Bio- metrics. Interindividual variability was modeled on clear-
analysis of plasma samples for 2-IB levels was performed ance, and a proportional residual error model was applied.
by validated liquid chromatography-mass spectrometry/mass Individual estimates for the PK profiles were obtained
spectrometry method (Charles River Labs, ‘s-Hertogen- using the population parameter estimates and random
bosch, the Netherlands). The lower limit of quantification effects. Through simulation of the predicted concentra-
(LLOQ) of 2-IB in human EDTA plasma was 5.00 ng/mL, tions the following exposure metrics were computed for
with a dynamic range of 5.00–5000 ng/mL. Between-run each patient:
and within-run coefficients of variation were < 20%. Samples
were stored at − 80 °C until analysis. 1. Cmax (observed maximum plasma concentration over the
PK analysis was performed using nonlinear mixed effects full treatment duration).
modeling (NONMEM; version 7.4; ICON Development 2. AUC​ 0–4h (area under the plasma concentration–time
Solutions, Ellicott City, MD, USA), with the main aim to curve from time 0 to 4 h after infusion).
obtain individual exposure metrics for 2-IB [15]. As the 3. AUC​ 0–∞ (area under the plasma concentration–time
dataset was very small (27 observations from 7 subjects), curve from time 0 to infinity).
the dataset was enriched with interim data after completion 4. Tmax (time at maximum plasma concentration).
of part A of the 2-STEP study (31 observations from 6 sub- 5. t½ (terminal elimination half-life).
jects), which became available during the execution of the 6. CL (clearance, model estimate).
current trial [13]. An overview of the main characteristics of 7. Vss (volume of distribution at steady state, model esti-
the 2-STEP and current study is provided in Table 1. mate).
Model development included testing of one- and two-
compartment models and the optimization of the interin-
dividual variability for the PK model parameter, as well as 2.8 Data Collection and Statistical Analysis
the residual variability. The model was subjected to a visual
predictive check. For this phase IIa study, no formal statistical analysis was
performed. Data were collected regarding the pregnancy,
delivery, and neonatal admission including postmen-
strual age, biometry, time of birth, Apgar at 5 min, time
of admission at the neonatal ward, and Thompson score
Table 1  Main characteristics of the model developmental dataset
at age 1–3 h. The aEEG (CFM Olympic Brainz Moni-
Parameter Study tor; Natus Medical Incorporated, Pleasanton, CA, USA)
2-STEP part A [13] TIBC was recorded during the first 48 h and analyzed using
visual interpretation of the background pattern [16] and
Dose (mg/kg) 0.16 0.16
presence of seizures on the raw EEG signal [17]. For the
Infusion duration (min) 1 1
assessment of safety and tolerability, vital signs, including
Treatment duration (h) 24 24
breathing and heart frequency, blood pressure, transcuta-
Number of doses administered 4 6
neous oxygen saturation, and temperature were monitored
Dose interval (h) 6 4
and recorded 15 min before, just before, 15 and 30 min
Number of samples 31 27
after the administration of the study drug, every hour until
Number of subjects 6 7
24 h after start of the study drug, and at 36 h and 48 h
Race Caucasian Black
for a maximum of 7 days using assessments done as part
Income country High Low
of the standard care. The clinical course of each patient
Body temperature Hypothermic Normothermic
was followed closely, and local tolerance and AEs/SAEs
TIBC Two-IminoBiotin in the Democratic Republic of Congo including mortality were monitored. Data are reported
2-Iminobiotin for Birth Asphyxia in Low-Income Countries 99

as mean ± SEM or as percentages, when appropriate. 3.2 Outcome


Not normally distributed data were reported as median
(range). The vital signs from admission until 48 h after HIE are
shown in Fig. 1. No relevant change in heart rate, non-
invasive blood pressure, transcutaneous oxygen saturation,
3 Results or spontaneously respiratory rate was observed over time.
At admission the mean rectal temperature of the patients
3.1 Patients was 34.9 ± 0.6 °C, increasing to a stable temperature of
35.9 ± 0.4 °C at 3 h after start of 2-IB treatment. No hypo-
Seven infants were included in the trial during the 5-month thermia was instituted.
study period. All mothers had prenatal consultations. No relevant effects on heart rate and blood pressure were
Three infants were admitted at the University Hospital of seen during and after administration of 2-IB. There was
Kinshasa, two at the Saint Joseph Hospital and another two no need for intervention due to respiratory and circulatory
at the Clinique Ngaliema, of which two patients were out- insufficiency from the start until 15 min after administration
born. Maternal and neonatal characteristics of the included of the study drug.
patients are shown in Table 2. All the infants needed resus- Three patients died: two after completing the study proto-
citation until 10 min after birth with facial mask and room col (patient 1 at 3 days after birth due to a neonatal infection
air. Blood gas analysis for pH and base excess was not and patient 4 at 2 days after birth due to resistant hypoglyce-
available in all hospitals. None of the patients had intra- mia with severe metabolic acidosis); patient 3 was moribund
uterine growth restriction, neither were there clinical signs at inclusion and died 1 h after inclusion, and should have
of chorioamnionitis. been excluded from this study (for vital signs, see Online
All the infants experienced HIE with a median Thomp- Resource 1 in the ESM). None of these (severe) adverse
son score of 10 (range 8–16) between 1 and 3 h after birth. events were judged by the investigators as being (potentially)
Clinical seizures were observed in three infants at the time related to the study drug.
of admission. All seven neonates had aEEG recordings. A continuous
normal voltage pattern was seen in four out of seven patients
at inclusion, which remained normal until 48 h after birth.
Table 2  Maternal and neonatal characteristics
One of these patients (patient 2) had repetitive seizures at
Maternal factors 36 h after inclusion and was treated with phenobarbitone
Maternal age, years 31 ± 2.8 with good effect. All these four patients had sleep–wake
Primiparity, n (%) 2 (29) cycling at 48 h after birth. Patient 1 had a burst suppression
Preeclampsia, n (%) 1 (14) pattern with seizures at admission that remained identical at
Urogenital infection, n (%) 0 (0) 48 h after birth, despite phenobarbitone treatment. Patient 3
Premature rupture of membranes, n (%) 2 (29) had a continuous low voltage pattern with seizures at admis-
Emergency caesarean section, n (%) 5 (71) sion, 1 h before death. Patient 4 had a discontinuous normal
Inborn, n (%) 5 (71) voltage with severe seizures at admission and evolved to
Neonatal factors continuous low voltage at 48 h after birth.
Male gender, n (%) 2 (29)
Postmenstrual age (weeks) 38.8 ± 0.7
Biometry 3.3 Pharmacokinetics
Birth weight (g) 3150 ± 189
Length (cm) 48.2 ± 1.1 Full PK data of 2-IB in six subjects were described using a
Head circumference (cm) 34.1 ± 0.8 two-compartment model based on a parameter estimation
Apgar score of the dataset combined with part A from the 2-STEP study.
1 min 3 (1–7) The final model was a two-compartment model similar to
5 min 4 (2–6) that of part A from the 2-STEP study with interindividual
10 min 6 (4–7) variability detected on clearance alone. In Fig. 2, the clear-
Thompson score between 1–3 h after birth 10 (8–16) ance estimates from the subjects in the model develop-
Duration of resuscitation (min) 14.8 ± 3.1 (unknown ment dataset are shown of the patients in the current TIBC
in 1 patient) study and group A of the 2-STEP study. Median clear-
Admission at neonatal ward (min) 49 ± 11 ance was 0.539 L/h in normothermic patients, treated with
Data are presented as mean ± standard error of mean or when appro- 0.16 mg/kg every 4 h for 24 h compared with a clearance
priate median (range) and number (percentage) of 0.468 L/h in hypothermic patients, treated with the same
100 T. Biselele et al.

Fig. 1  Vital parameters a heart


rate, respiratory rate and trans-
a 180 100
cutaneous oxygen saturation, 160 90
b systolic and diastolic blood 80
pressure, and c temperature, 140
from admission until 48 h after 70

Saturaon (%)
120

Rate (min-1)
start of 2-iminobiotin treatment. 60
100
Data is mean ± SD. X-axis is 50
not linear 80
40
60
30
40 20
20 10
0 0

heart rate respiratory rate oxygen saturaon

b 90
80
Blood pressure (mm Hg)

70
60
50
40
30
20
10
0

systolic BP diastolic BP
c
38.0
Temperature (°C)

37.0
36.0
35.0
34.0
33.0
T= 12H 30MIN
T= -15 MIN
T= 0H 15MIN
T= 1H
T= 3H
T= 3H 59MIN
T= 4H 30MIN
T= 6H
T= 7H 45MIN

T= 9H

T= 11H 59MIN

T= 14H
T= 15H 45MIN
T= 16H 15MIN
T= 17H
T= 19H
T= 19H 59MIN
T= 20H 30MIN
T= 22H
T= 24H
T= 36H
T= 48H
T= 8H 15 MIN

T= 11H

temperature

.
Data is mean ± SD X-axis is not linear.

dose every 6 h for 24 h. The residual variability was modeled little to no accumulation of 2-IB, except for patient 1 who was
proportionally. hypothermic from 13 to 48 h after inclusion and was shown to
The goodness-of-fit plots are given in Online Resource have a neonatal infection afterwards (Fig. 3).
2 (see ESM). A visual predictive check is given for the two Individual exposure metrics based on the individual model
studies that were included in the model development dataset parameter estimates are described in Table 4. Median AUC​
separately (Online Resource 3, see ESM). The model was able 0–4h was 664 ng*h/mL (range 414–917), nearly twice as high
to accurately predict both the medians as well as the variability as targeted. The median clearance of 2-IB treated patients
of the observations for both the 2-STEP and the current study was 0.539 L/h. There was no apparent relationship observed
as given by the precision in both the predicted and observed between clearance and body weight, gestational age, and dis-
5th and 95th percentiles. Model parameter estimates are given ease severity, defined as Thompson score at admission, in this
in Table 3. Individual plots show high peak concentrations and group of patients (Online Resource 4, see ESM).
2-Iminobiotin for Birth Asphyxia in Low-Income Countries 101

due to respiratory and circulatory insufficiency during/after


2-IB administration. In this study, three patients died, of
which one patient was moribund at inclusion with a Thomp-
son score of 16 and should not have been included. This is
comparable with the mortality rate (44%) in the study that
was conducted in the same hospitals to investigate the fea-
sibility of neuroprotective treatment in the DRC [18]. None
of these SAEs were judged by the investigators and DSMB
as being (potentially) related to the study drug, but were
caused by neonatal infection (patient 1) at 3 days after birth,
and resistant hypoglycemia and severe metabolic acidosis
(patient 4) at 2 days after birth. Although only seven patients
were included in this trial, no 2-IB-related side effects were
reported by the principal investigator experienced in treat-
ing patients with moderate to severe HIE in LIC. In total, 25
neonates with HIE have now been treated in the pilot study
(n = 6), the 2-STEP study (n = 12), and the current TIBC
study (n = 7). Although no formal adverse drug reaction
Fig. 2  Boxplot for individual clearance determined using the cur-
rent model for individuals participating in the 2-STEP study cohort probability scale was performed in this or the other studies,
A and in the current study. The horizontal line indicates the median, no time-related reactions to the study drug infusion were
the grey area the interquartile range and the whiskers the range of the found in any of the studies.
data. Clearance of patients of the 2-STEP study [13] cohort A, treated
The present dosing regimen resulted in an AUC nearly
with a dose of 0.16 mg/kg/dose every 6 h for 24 h during hypother-
mia, and clearance of patients in current study, treated with a similar twice as high as that targeted based on the piglet model [6].
dose every 4 h for 24 h during normothermia. Data from patient 3 The discrepancy between expected versus observed AUC
of the current study have been excluded since this patient should not in this population is in all likelihood caused by the fact that
have been included
the pilot data was based on only two subjects. The vari-
ability in clearance could not be explained by bodyweight,
Table 3  Pharmacokinetic model parameters of 2-IB from 2-STEP gestational age, or Thompson score at admission (Online
and TIBC study Resource 4, see ESM). As serum creatinine concentrations
Parameter Estimate (%SE) 95% CI during the first few days of life are confounded by maternal
transfer, determination of individual renal function based on
Structural model parameters serum creatinine is subject to a high degree of uncertainty
CL (L/h) 0.435 0.303–0.567 in neonates, and is not part of the standard care in an LIC.
Vc (L) 0.740 0.467–1.01 Moreover, the present study was a pilot study and not pow-
Q (L/h) 2.12 1.69–2.56 ered for statistical inference.
Vp (L) 1.07 0.638–1.50 The current study leads to a better understanding of the
Interindividual variability PK of 2-IB in normothermic neonates. In future, larger scale
CL (%) 48 (31) 31–61 trials will allow the development of a model that is more pre-
Residual variability cise and less sensitive to potential bias so that the presence
Proportional 36 (25) 26–44 of clinically relevant covariates can be determined, thereby
CI confidence interval, CL clearance, Q intercompartmental clear- optimizing the dosing schedule of 2-IB.
ance, SE standard error of estimate, Vc volume of the central com- For a randomized study aimed at looking at effectivity
partment, Vp volume of the peripheral compartment of 2-IB after moderate to severe HIE in LIC, the dose regi-
men has to be adjusted to meet the AUC​0–4h target criteria
of 365 ng*h/mL. To achieve this, either the dose can be
4 Discussion reduced to 0.09 mg/kg per dose with 4-hourly intervals or
the dose interval can be prolonged to 6-hourly intervals with
This is the first PK and safety study of 2-IB in newborns a dose reduction to 0.14 mg/kg.
with moderate to severe hypoxia-ischemia in a low-resource In order to administer the neuroprotective treatment
setting, such as in the DRC. within the therapeutic window, the baby has to be trans-
Regarding safety, during and until 30 min after 2-IB ported to a neonatal ward facility within 6 h of birth. In LIC
administration, no relevant differences in vital parameters such as sub-Saharan Africa, this is a challenge, since there is
were detected. There was no need for clinical intervention a lack of good public transport and communications systems,
102 T. Biselele et al.

Fig. 3  Pharmacokinetic plots of 2-iminobiotin (2-IB) for each indi- individual model fit, and black dashed lines the population model fit.
vidual patient. Grey lines indicate the nominal dosing times, red dots Time is the time after first administration of 2-IB. Patient 3 is omitted
are the observed 2-IB plasma concentrations. Blue lines represent the since he died 1 h after birth

Table 4  Exposure metrics for individual patients of current TIBC study

Patient BW (kg) Cmax (ng/mL) AUC​0–4h (ng*h/mL) AUC​inf (ng*h/mL) Tmax (h) t½ (h) CL (L/h) Vss (L)

1 2.95 387 863 2284 21.0 6.28 0.207 0.74


2 3.10 167 534 676 0.1 1.93 0.734 0.74
4 3.40 407 917 1958 21.0 4.72 0.278 0.74
5 2.30 NA 414 538 19.9 2.05 0.685 0.74
6 2.90 359 691 1183 21.0 3.41 0.392 0.74
7 3.75 56 637 800 21.0 1.89 0.750 0.74
Median 3.03 359 664 992 21.0 2.73 0.539 0.74
Range 2.3–3.75 56–407 414–917 538–2284 0.1–21.0 1.89–6.28 0.207–0.750 0.74–0.74

AUC​ area under the curve, BW birth weight, CL clearance, Cmax maximal serum concentration, inf infinity, NA not available, t½ half-life, Tmax
time to maximal serum concentration, Vss volume of distribution at steady state model estimate
2-Iminobiotin for Birth Asphyxia in Low-Income Countries 103

and many patients cannot afford private transport. However, Conflict of interest Authors T. Biselele, J. Bambi, D. Betukumesu, Y.
in the setting of Kinshasa, we showed earlier that about 45% Ndiyo, G. Tabu, J. Kapinga, V. Bola, P. Makaya, and P. Vis have no dis-
closures. Authors H. Tjabbes and C. Peeters-Scholte are co-founders,
of the patients are able to reach the hospital in time [18]. In consultants for and shareholder of Neurophyxia BV. Author H. Tjabbes
this study, five out of seven patients were inborn, and the is Vice-President Regulatory and Medical Affairs of Neurophyxia, and
mean time to admission was 49 min. author C. Peeters-Scholte is Chief Scientific Officer.
Several neuroprotective treatment strategies are currently
Informed consent All procedures performed in studies involving
being investigated to reduce cerebral damage after HIE. TH human participants were in accordance with the ethical standards of
has become the standard of care for neonatal encephalopa- the ethical committee of the Public Health School of the University
thy in high-income countries [19]. However, a meta-analysis of Kinshasa (DR Congo) and the ethical committee of the Univer-
on TH in LIC using low-cost cooling techniques did not sity of Utrecht (The Netherlands), and in line with the 1964 Helsinki
declaration and its later amendments or comparable ethical standards.
show a significant reduction in neonatal mortality [3]. It was Written informed consent was obtained from one of the parents in
concluded that adequately randomized controlled trials are local language (Lingala) or French before start of the study. When the
required before cooling can be offered in LIC [20]. consenting parent was illiterate, informed consent was given verbally
Pharmacological strategies to reduce cerebral brain injury in the presence of an impartial witness.
following perinatal asphyxia in LIC should be affordable,
cost effective, and easy to be administered. Also, storage
conditions have to be compatible with a high-temperature
environment, since power shutdowns often occur in LIC. References
2-IB proved to be a very stable drug that can be stored for
1. United Nations Inter-agency Group for Child Mortality Estima-
at least 6 months at temperatures up to 40 °C. It can be tion. In: Levels and trends in child mortality. Report 2018. https​
easily administered either through an umbilical catheter or ://data.unice​f.org/wp-conte​nt/uploa​ds/2018/10/Child​-Morta​lity-
a peripheral infusion, has a predictable PK profile, and is Repor​t-2018.pdf. Accessed 13 Nov 2019.
2. Lawn JE, Gravett MG, Nunes TM, Rubens CE, Stanton C, Group
affordable.
GR. Global report on preterm birth and stillbirth (1 of 7): defini-
Based on the concepts of value-based healthcare, it is tions, description of the burden and opportunities to improve data.
important to perform a cost-benefit analysis for the addi- BMC Pregnancy Childbirth. 2010;10(Suppl 1):S1.
tive treatment with 2-IB after birth asphyxia. In 2017, about 3. Pauliah SS, Shankaran S, Wade A, Cady EB, Thayyil S. Therapeu-
tic hypothermia for neonatal encephalopathy in low- and middle-
2.3% of the global burden of disease was being caused by
income countries: a systematic review and meta-analysis. PLoS
encephalopathy due to birth asphyxia and trauma [21]. For One. 2013;8(3):e58834.
TH in the Western world, a number needed to treat of seven 4. Albrecht M, Zitta K, Groenendaal F, van Bel F, Peeters-Scholte C.
was found [19]. Although the efficacy of 2-IB still needs to Neuroprotective strategies following perinatal hypoxia-ischemia:
taking aim at NOS. Free Radic Biol Med. 2019;142:123–31.
be tested in a future randomized clinical trial, the production
5. Peeters-Scholte C, Koster J, Veldhuis W, van den Tweel E, Zhu C,
and administration costs of 2-IB are low, making a potential Kops N, et al. Neuroprotection by selective nitric oxide synthase
treatment with 2-IB even in LIC feasible. inhibition at 24 hours after perinatal hypoxia-ischemia. Stroke.
2002;33(9):2304–10.
6. Bjorkman ST, Ireland Z, Fan X, van der Wal WM, Roes KC,
Colditz PB, et al. Short-term dose-response characteristics of
5 Conclusion 2-iminobiotin immediately postinsult in the neonatal piglet after
hypoxia-ischemia. Stroke. 2013;44(3):809–11.
7. van den Tweel ER, van Bel F, Kavelaars A, Peeters-Scholte CM,
This is the first study investigating safety and PK of 2-IB
Haumann J, Nijboer CH, et al. Long-term neuroprotection with
after moderate to severe HIE in LIC. The present dosing 2-iminobiotin, an inhibitor of neuronal and inducible nitric oxide
regimen resulted in higher AUC levels than targeted. The synthase, after cerebral hypoxia-ischemia in neonatal rats. J Cereb
dose regimen will be adjusted to target the AUC in future Blood Flow Metab. 2005;25(1):67–74.
8. Nijboer CH, Groenendaal F, Kavelaars A, Hagberg HH, van Bel
efficacy trials, which will inform improved PK models. No
F, Heijnen CJ. Gender-specific neuroprotection by 2-iminobiotin
adverse effects that could be attributed to the use of 2-IB after hypoxia-ischemia in the neonatal rat via a nitric oxide inde-
were observed. pendent pathway. J Cereb Blood Flow Metab. 2007;27(2):282–92.
9. Neurophyxia. In: Literature: phase 1 clinical study: first-in-man
Acknowledgements We greatly thank Laurent Favié and Floris 2-IB dose escalation study. https​://www.neuro​phyxi​a.com/files​/
Groenendaal, the investigators of the 2-STEP study, who included resul​ts-phase​1-clini​cal-trial​.pdf. Accessed 13 Nov 2019.
patients in the 2-STEP study and provided PK data of patients from 10. Neurophyxia. In: Literature: determination of target dose for
Group A to enrich the PK population model. phase II study of 2-iminobiotin (2-IB) in neonates with perinatal
asphyxia. https​://www.neuro​phyxi​a.com/files​/2012-01-18-Deter​
minat​ion-of-targe​t-dose-for-Phase​-II.pdf. Accessed 13 Nov 2019.
Compliance with Ethical Standards 11. https​://www.clini​caltr ​ialsr​egist​er.eu. Accessed 13 Nov 2019.
Eudra CT number 2011-002502-74.
Funding This study was funded by Neurophyxia BV. 12. Holford N, Heo Y-A, Anderson B. A pharmacokinetic standard
for babies and adults. J Pharm Sci. 2013;102:2941–52.
104 T. Biselele et al.

13. Favié LMA, Peeters-Scholte CMPCD, Bakker A, Tjabbes H, 17. Hellström-Westas L. Amplitude-integrated electroencephalogra-
Egberts TCG, van Bel F, Rademaker CMA, Vis P, Groenendaal phy for seizure detection in newborn infants. Semin Fetal Neona-
F. Pharmacokinetics and short-term safety of the selective NOS tal Med. 2018;23(3):175–82.
inhibitor 2-iminobiotin in asphyxiated neonates treated with thera- 18. Biselele T, Bambi J, Naulaers G, Tabu G, Kapinga J, Bola V, et al.
peutic hypothermia. Pediatr Res. 2019. https​://doi.org/10.1038/ Observational study shows that it is feasible to provide neuro-
s4139​0-019-0587-1. protective treatment for neonatal encephalopathy in low-income
14. Thompson CM, Puterman AS, Linley LL, Hann FM, van der Elst countries. Acta Paediatr. 2018;107(8):1345–9.
CW, Molteno CD, Malan AF. The value of a scoring system for 19. Jacobs SE, Berg M, Hunt R, Tarnow-Mordi WO, Inder TE, Davis
hypoxic ischaemic encephalopathy in predicting neurodevelop- PG. Cooling for newborns with hypoxic ischaemic encephalopa-
mental outcome. Acta Paediatr. 1997;86:757–61. thy. Cochrane Database Syst Rev. 2013;1:CD003311.
15. Beal S, Sheiner LB, Boeckmann A, Bauer RJ. NONMEM user’s 20. Montaldo P, Pauliah SS, Lally PJ, Olson L, Thayyil S. Cooling
guides (1989–2011). Ellicott City: Icon Development Solutions; in a low-resource environment: lost in translation. Semin Fetal
2011. Neonatal Med. 2015;20(2):72–9.
16. Hellström-Westas L, Rosén I. Continuous brain-function monitor- 21. IHME. Global burden of disease. https​://vizhu​b.healt​hdata​.org/
ing: state of the art in clinical practice. Semin Fetal Neonatal Med. gbd-compa​re/. Accessed 7 Nov 2019.
2006;11(6):503–11.
Reproduced with permission of copyright owner. Further reproduction
prohibited without permission.

You might also like