Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

European Journal of Pharmaceutical Sciences 141 (2020) 105134

Contents lists available at ScienceDirect

European Journal of Pharmaceutical Sciences


journal homepage: www.elsevier.com/locate/ejps

Safety, tolerability, and pharmacokinetics of adamgammadex sodium, a T


novel agent to reverse the action of rocuronium and vecuronium, in healthy
volunteers
YingYing Jianga,b,1, YuJun Zhanga,b,c,1, ShunJu Xianga, WenLing Zhaoa,b,c, Jin Liua,b,c, ,

WenSheng Zhanga,b,c,

a
Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University,
Chengdu, Sichuan, China
b
National-Local Joint Engineering Research Center of Translational Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
c
Sichuan Engineering Laboratory of Transformation Medicine of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China

ARTICLE INFO ABSTRACT

Keywords: Neuromuscular blockers (NMBs) selectively block neuromuscular transmission at the N2-nicotinic receptor on
Neuromuscular block antagonists motor neurons to paralyze skeletal muscles, and are mainly used to facilitate tracheal intubation and surgical
phase I study procedures. Rapid reversal is necessary in clinical practice to avoid profound block and reduce recovery time.
dose escalation Adamgammadex sodium is a modified γ-cyclodextrin derivative consisting of a lipophilic core and a hydrophilic
pharmacokinetics
outer end that forms an inactive tight inclusion complex with free molecules of rocuronium and vecuronium. In
adamgammadex sodium
preclinical study, adamgammadex produced a concentration-dependent reversion effect of neuromuscular
blockade induced by rocuronium in beagle dogs. Furthermore, adamgammadex had a less potential side effects
than sugammadex and other clinical used neuromuscular block antagonists. In this study, the objective was to
assess the safety, tolerability, and pharmacokinetics of single intravenous injection of adamgammadex in healthy
volunteers. Approved by the China Food and Drug Administration, 52 healthy volunteers (half male and half
female) were enrolled in this single-center, randomized, double-blind placebo-controlled study. No serious ad-
verse effects were happened in this study. The overall frequency of adverse effects in adamgammadex was
similar for that in placebo, and there was no specific adverse effect in adamgammadex. All of the volunteers
bearing the adverse effects were recovered to normal without any treatment or intervention. In pharmacokinetic
study, the value of half-time, Tmax, and clearance were not changed significantly, and the Cmax and AUC0–∞
increased with a similar ratio of the escalating doses. For dose proportionality analysis of adamgammadex, the
estimate of slope was close to 1, and it was not significantly different from 1 after doses (AUC0-∞, 0.9965
[90%CI, 0.9468, 1.046]; Cmax, 0.9462 [90%CI, 0.8800, 1.012]). Therefore, adamgammadex exposure in plasma
increased in a dose- proportional manner. The urinary route is a significant excretory pathway for adamgam-
madex, and it is mostly completed at 8 h. All the results in this study showed that adamgammadex may be a
novel safe neuromuscular blockade reversal agent .

1. Introduction reduce recovery time. The traditional neuromuscular block antagonists


neostigmine and edrophonium are anticholinesterase drugs, which in-
Neuromuscular blockers (NMBs) selectively block neuromuscular hibit cholinesterase to reduce the degradation of acetylcholine
transmission at the N2-nicotinic receptor on motor neurons to paralyze (Singh and Sadiq, 2019). However, they may cause side effects such as
skeletal muscles, and are mainly used to facilitate tracheal intubation bradycardia, salivation, bronchospasm, and vomiting due to their ef-
and surgical procedures (Murphy, 2018; Murphy et al., 2004). Rapid fects on muscarinic acetylcholine receptors and nicotinic receptors in
reversal is necessary in clinical practice to avoid profound block and other tissues. In order to avoid these, cholinergic receptor antagonists

Corresponding authors at: Laboratory of Anesthesia and Critical Care Medicine, Department of Anesthesiology, Translational Neuroscience Center, West China

Hospital, Sichuan University, Chengdu, Sichuan, China


E-mail addresses: scujinliu@gmail.com (J. Liu), zhang_ws@scu.edu.cn (W. Zhang).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.ejps.2019.105134
Received 27 July 2019; Received in revised form 14 October 2019; Accepted 29 October 2019
Available online 31 October 2019
0928-0987/ © 2019 Elsevier B.V. All rights reserved.
Y. Jiang, et al. European Journal of Pharmaceutical Sciences 141 (2020) 105134

are usually administered in combination with atropine, but it them- Table 1


selves are also associated with side effects such as tachycardia, dry Number of subjects treated in each dose group.
mouth, and blurred vision. Meanwhile, traditional neuromuscular Adamgammadex dose group (mg/kg)
blockade antagonists have a limited effect against profound block. 0.5 2 4 8 16 24 32
Sugammadex sodium (Bridion®, Merck & Co., Inc.) is approved to
No. of subjects receiving 2 6 6 6 6 6 6
reverse muscle relaxation by rocuronium bromide in more than 80
active
countries (Groudine et al., 2007; Sacan et al., 2007; Shields et al., 2006; No. of subjects receiving 2 2 2 2 2 2 2
Sorgenfrei et al., 2006; Sparr et al., 2007; Suy et al., 2007; placebo
Vanacker et al., 2007). However, it has been rejected three times by the Total subjects 4 8 8 8 8 8 8
U.S. Food and Drug Administration (FDA) in 2008, 2013, and 2015, due
to its potential allergic reactions, and it also carries a risk of post-
operative bleeding. It was finally approved with a caution against hy- the maximum tolerance dose (MTD) in rats was more than 3 g/kg. Due
persensitivity reactions and anaphylaxis (Baldo and McDonnell, 2014; to the linear relationship of pharmacokinetic features with the dosage
Takazawa et al., 2014; Tsur and Kalansky, 2014). Furthermore, su- in rats and beagle dogs, we set the highest dose of adamgammadex at
gammadex also increased the risk of postoperative bleeding 32 mg/kg.
(Carron, 2015; Rahe-Meyer et al., 2014). Based on this range, we tested seven escalating doses of 0.5, 2, 4, 8,
Adamgammadex sodium acts similarly to sugammadex, with pre- 16, 24, and 32 mg/kg. Each subject remained in the clinical unit from
clinical studies demonstrating similar efficacy and fewer potential side the day before receiving adamgammadex or placebo until the day after
effects (Chen et al., 2015; Qi et al., 2018). It consists of a modified γ- dosing. Follow-up visits were made 4 and 8 days after dosing. Prior to
cyclodextrin derivative with a lipophilic core and a hydrophilic outer beginning administration, one catheter was inserted into the median
end that forms an inactive tight inclusion complex with free molecules cubital veins of the left and right arms for drug administration and
of rocuronium and vecuronium. In beagle dogs, adamgammadex pro- blood sample collection, respectively. Subjects were administered
duces a concentration-dependent reversion effect of neuromuscular adamgammadex diluted with saline to 125 mg/mL or placebo (saline
blockade induced by rocuronium. In zebrafish, it has been found to only) i.v. at 1 mL/s. After administration, an interim safety report was
modulate tryptase, suggesting a lower potential for causing hy- made depending on the common terminology criteria (CTCAE, version
persensitivity and anaphylaxis. Finally, it shows negligible detrimental 4.03) for adverse events (AEs), including the results of all safety as-
effects on cardiac function and does not pose a risk of bleeding. sessments during that period. The intensity of AEs was classified into
Adamgammadex has been approved by the China FDA (approval five categories according to the CTCAE version 4.03: grade 1, mild
number, 2015L05410) for clinical trials in reversing the neuromuscular (asymptomatic or mild symptoms, intervention not indicated); grade 2,
blockade effect induced by rocuronium and vecuronium. In this study, moderate (minimal, local or noninvasive intervention indicated); grade
we aimed to assess the safety, tolerability, and pharmacokinetics of 3, severe (medically significant but not immediately life-threatening);
single intravenous injection of adamgammadex sodium in healthy vo- grade 4, serious (life-threatening consequences, urgent intervention
lunteers. indicated); and grade 5, death related to AEs. The last two were clas-
sified as serious AEs.
2. Materials and methods Groups were tested in succession with the lowest dose group tested
first. Subsequent groups receiving increased doses were tested only if
2.1. Subject recruitment the agent was found to be safe and well-tolerated in the previous
groups. Testing was suspended when more than 50% of treated subjects
Approved by the China FDA, fifty-two healthy volunteers (half male in a dose group showed drug-related AEs of level 2 or above.
and half female) were enrolled in this single-center, randomized,
double-blind, placebo-controlled study at West China Hospital, Sichuan 2.3. Safety assessment
University, Chengdu, China (registration number,
ChiCTR1900024437). Only healthy young male and female subjects Physical examinations were performed at 24 h post-dose. Basic vital
aged 18–40 with body mass index (BMI) of 19–26 kg/m2 and weight of signs including blood pressure, breathing rate, heart rate, and tem-
50–90 kg for males and 45–85 kg for females were included. The sub- perature were measured prior to dosing and 2, 4, 8, 12, and 24 h after
jects with diagnosed digestive, urinary, cardiovascular, circulatory, dosing. Twelve-lead ECG was monitored continuously from 10 min pre-
respiratory, neuropsychiatric, endocrine, or and metabolic diseases dose to 40 min post-dose, and it were gathered at prior to dosing, 15 s,
were ineligible for enrollment. The subjects were also excluded if they 2, 3, 4, 5, 6, 8, 10, 15, 25, 40 min, 1, 1.5, 2, 3, 4, 6, 8, 10, and 24 h after
had congenital hemorrhagic diseases, coagulative dysfunction, history dosing. Coagulative function was measured 30 min before dosing and
of allergic reactions (e.g., food allergies, drug allergies, specific allergic again at 3, 10, 25, and 40 min, and at 1, 3, and 24 h post-dose. These
reactions, or asthma attacks), sensitivity to cyclodextrin or other drugs, examinations were repeated at follow-up visits. Signs of anaphylaxis
recent febrile disease (past 7 days), previous use of sugammadex, or were noted at 24 h post-dose, including erythema, urticaria, angioe-
abnormalities in X-ray chest radiography, 12-lead electrocardiographic dema, hypotension, bronchospasm, and asthma (Brown, 2004). At 48 h
(ECG), blood biochemistry, urine biochemistry, serum virology, or post-dose, we further evaluated hyperpyrexia, lymphadenectasis, er-
blood clotting function tests ythema, papules, pustules (acute generalized eruptive pustulosis),
blisters or blebs (epidermolysis bullosa), liver injury, eosinophilia, and
2.2. Treatment neutrophilia. Related clinical symptoms of anaphylaxis were evaluated
during the follow-up visit. Blood trypsin levels were tested in subjects
Eligible subjects were randomized to receive placebo or adam- with anaphylaxis within 3 h of onset.
gammadex (Table 1). Based on the previous preclinical pharmacolo-
gical and toxicological studies, the no-observed-adverse-effect level 2.4. Pharmacokinetic study
(NOAEL) of adamgammadex after 15-days repeated i.v. administration
was 200 mg/kg in rats (transfer to human equivalent dose, 32.3 mg/kg) Venous blood samples were collected at 0, 2, 3, 4, 5, 6, 8, 10, 15, 25,
and 450 mg/kg in beagle dogs (transfer to human equivalent dose, and 40 min, and at 1, 1.5, 2, 3, 4, 6, 8, 10, and 24 h after administration
250 mg/kg). The minimum dose in this phase I escalating-dose trial, to quantify the concentration of adamgammadex in plasma. Blood
0.5 mg/kg, was set as one-sixth of the NOAEL dose in rats. Meanwhile, samples were also collected at the same for subjects receiving the

2
Y. Jiang, et al. European Journal of Pharmaceutical Sciences 141 (2020) 105134

placebo. Blood samples (4 mL) were transferred to anticoagulant-coated Table 3


tubes and centrifuged at 1500–2000 g and 2–8 °C within 15 min to The summary of all adverse events (AEs) in this study.
obtain plasma, which was transferred into two sample tubes and stored Dose group Placebo Adamgammadex
at −70 °C. Urine samples were collected prior to dosing and at 0–2, No. of subjects No. of AEs No. of subjects No. of AEs
2–4, 4–6, 6–8, 8–10, 10–12, and 12–24 h post-dose. Timing and volume
0.5 mg/kg 2 (100%) 5 1 (50.0%) 1
of urine were recorded at each time point and two 1 mL samples were
2 mg/kg 1 (50.0%) 1 4 (66.7%) 7
retained and stored at −20 °C. 4 mg/kg 0 0 1 (16.7%) 2
Concentration of adamgammadex in plasma and urine were de- 8 mg/kg 1 (50.0%) 1 3 (50.0%) 5
termined at Q2 Solutions Co., Ltd. (Beijing, China) according to liquid 16 mg/kg 1 (50.0%) 1 2 (33.3%) 2
chromatography-mass spectrometry (LC/MS) methods developed and 24 mg/kg 1 (50.0%) 1 5 (83.3%) 9
32 mg/kg 2 (100%) 3 4 (66.7%) 6
validated by the China FDA (Pharmacopoeia, 2015) and the U.S. FDA
Total 8 (57.1%) 12 20 (52.6%) 32
(FDA, 2001; Viswanathan et al., 2007). The LC/MS system consisted of
an API 5500 mass spectrometer (AB Sciex) and Waters Acquity UPLC
system equipped with the Acquity UPLC BEH C18 Column (1.7 μm,
2.1 mm × 50 mm). Separation was carried out using 0.05% ammonium each group, the ratio of male and female was 1:1, mean age was
hydroxide in water as mobile solvent A and methanol/acetonitrile, the 24.2 ± 2.9 years old, mean body height was 1.64 ± 0.08 m, baseline
solvent B, at a volume ratio of 50:50, at an initial flow rate of 200 μL/ weight was 59.21 ± 9.52 kg, and BMI was 21.79 ± 2.00 kg/m2. Three
min at 40 °C with a TurboIonSpray™ in negative mode and selected subjects had a history of smoking and none had a history of alcohol
reaction monitoring (SRM) scanning. A selected reaction monitoring abuse, drug abuse, allergy, or blood donation within the previous 3
mode at transitions of m/z 613.3 → 128.0 for adamgammadex and m/z months.
327.0 → 157.0 for the internal standard (labetalol) was used for
quantification with negative electrospray ionization interface. 3.1. Safety

2.5. Data analysis There were no serious AEs, and no subjects withdrew from the trial
due to AEs. Among those receiving adamgammadex, 20 subjects
The pharmacokinetic analysis and inferential analysis of pharma- (52.6%) experienced a total of 32 AEs (Table 3). In the placebo group,
cokinetic parameters were accomplished by IQVIA™ (Shanghai, China). eight subjects (57.1%) experienced a total of 12 AEs. Three cases of
Pharmacokinetic parameters were calculated by non-compartmental clinical symptoms or disease were observed in those receiving adam-
model using Phoenix WinNonlin® (version 6.4, NJ, USA). For the cal- gammadex (dizziness, cough, and upper respiratory infection), which
culation purpose, pre-dose samples that were below the lower limit of were judged by the investigator as non-related to the drug. Among
quantitation (LLOQ) or missing were assigned a numerical value of those receiving adamgammadex, clinical laboratory examination in-
zero. Parameters of pharmacokinetic in this study were as follows: the dicated increased blood ketone bodies in 15.8% of subjects, increased
value of maximum concentration (Cmax) and the time to acquire (Tmax); triglycerides in 7.9%, increased β2-microglobulin in 7.9%, decreased
the elimination half-time (t1/2); area under curve (AUC) from zero to fibrinogen in 5.3%, increased urinary glucose in 5.3%, increased ur-
the last time point (AUC0–t), from zero to 24 h post-dose (AUC0–24) and inary ketone bodies in 5.3%, increased urobilinogen in 5.3%, and he-
from zero to infinity (AUC0–∞), clearance (CL), volume of distribution maturia in 5.3%. In those receiving placebo, the incidence rate of ur-
(Vd), and apparent terminal rate constant (Kel). The following phar- inary ketone bodies increased in 28.6%, urinary glucose increased in
macokinetic parameters in urine were estimated: cumulative amount of 14.3%, triglycerides increased in 7.1%, thrombin time increased in
drug excreted in urine from zero to 24 h post-dose (Ae(0–24)), cumula- 7.1%, fibrinogen decreased in 7.1%, activated partial thromboplastin
tive fraction of dose excreted unchanged in urine from zero to 24 h post- time increased in 7.1%, urinary ketone bodies increased in 7.1%, and
dose (fe(0–24)), and renal clearance (CLr = Ae(0–24)/AUC0–24). The dose white blood cells were found in 7.1%.
proportionality analysis was calculated according to the equation A total of 11 AEs in eight subjects were judged as possibly related to
below: adamgammadex (Table 4). These included seven cases of increased
blood ketone bodies in (15.8% of subjects), two cases of increased ur-
The linear model: log(Y ) = intercept + slope × log(dose)
inary glucose (5.3% of subjects), and two cases of increased urinary
Exponentiation of both sides:Y = exp(intercept) × doseslope ketone bodies (5.3% of subjects). Among those receiving placebo, eight
potentially related AEs occurred, including four cases of increased
blood ketone bodies (28.6% of subjects), two cases of increased urinary
3. Results glucose (14.3% of subjects), one case of increased urinary ketone bodies
(7.1% of subjects), and one case of prolonged thrombin time (7.1% of
Of the 65 subjects signed the informed consent, 38 received subjects; Table 4).
adamgammadex and 14 received placebo. The number of subjects in Overall, adamgammadex-induced AEs had a lower incidence rate
each dose group and their demographics and other baseline char- than placebo, including increased blood ketone bodies (15.8% vs.
acteristics are shown in Tables 1 and 2. There were no premature dis- 28.6%), increased urinary glucose (5.3% vs. 14.3%), and prolonged
continuations from the study, and all subjects completed the trial. In thrombin time (0% vs.7.1%). There were no AEs specific to

Table 2
Characteristics of subjects (mean ± SD).
0.5 2 4 8 16 24 32
mg/kg mg/kg mg/kg mg/kg mg/kg mg/kg mg/kg

Age, yr 27.0 ± 2.5 21.6 ± 1.5 23.1 ± 3.6 25.4 ± 3.9 24.9 ± 1.7 25.0 ± 1.5 23.9 ± 2.6
Sex, M&F 2&2 4&4 4&4 4&4 4&4 4&4 4&4
Height, cm 166 ± 3 164 ± 6 161 ± 8 165 ± 11 165 ± 10 166 ± 9 164 ± 9
Weight, kg 59.0 ± 5.9 57.0 ± 8.3 55.9 ± 10.8 59.4 ± 9.9 60.8 ± 13.1 61.6 ± 8.6 60.6 ± 9.4

3
Y. Jiang, et al. European Journal of Pharmaceutical Sciences 141 (2020) 105134

Recovery
Recovery
Recovery
Recovery
Recovery
Recovery
Recovery
Recovery
Recovery
Recovery
Recovery
Outcome
Treatment

None
None
None
None
None
None
None
None
a
Intensity

I
I
I
I
I
I
I
I
Serum β-hydroxybutyrate ↑ (0.38 mmol/L, [0.02–0.27 mmol/L])

Serum β-hydroxybutyrate ↑ (0.42 mmol/L, [0.02–0.27 mmol/L])


Serum β-hydroxybutyrate ↑ (0.36 mmol/L, [0.02–0.27 mmol/L])

Serum β-hydroxybutyrate ↑ (0.37 mmol/L, [0.02–0.27 mmol/L])


Urine ketone body ↑ (2.0 (1+) mmol/L, [-])
Urinary glucose ↑ (8.3 (2+) mmol/L, [-])

Urinary glucose ↑ (3.9 (1+) mmol/L, [-])


Adverse event (value, [normal range])

Thrombin time ↑ (23.5 s, [14 – 22 s])

Fig. 1. The mean plasma concentration-versus-time curves (A) and the cumu-
lative excretion in urine (B) in healthy volunteers receiving single doses of
adamgammadex sodium (2–32 mg/kg).
Placebo group

0.5 mg/kg
0.5 mg/kg
0.5 mg/kg
0.5 mg/kg

16 mg/kg
24 mg/kg
32 mg/kg
2 mg/kg

adamgammadex. All subjects experiencing AEs recovered without any


Dose

treatment or intervention. No changes in basic vital signs or clinically


meaningful ECG abnormalities were observed. Summarily, the fre-
Recovery
Recovery
Recovery
Recovery
Recovery
Recovery
Recovery
Recovery
Recovery
Recovery
Recovery
Outcome

quency and categories of AEs did not differ significantly among the
doses tested, nor from placebo. These findings indicate that adam-
gammadex was well tolerated.
Treatment

Reference to the common terminology criteria for adverse events (CTCAE, version 4.03).
Adverse events judged as possibly related to administration of adamgammadex and placebo.

3.2. Pharmacokinetics
None
None
None
None
None
None
None
None
None
None
None

Following a single intravenous injection of 2, 4, 8, 16, 24, or 32 mg/


a

kg adamgammadex, mean plasma concentrations declined rapidly, with


Intensity

t1/2 calculated as 1.6–2 h (Fig. 1A, Table 5). Plasma concentrations fell
below LLOQ after 10 h for the 2, 4, and 8 mg/kg dose groups and after
I
I
I
I
I
I
I
I
I
I
I

14 h for the 16, 24, and 32 mg/kg dose groups. Tmax was from 0.050 to
Serum β-hydroxybutyrate ↑ (0.35 mmol/L, [0.02–0.27 mmol/L])
Serum β-hydroxybutyrate ↑ (0.42 mmol/L, [0.02–0.27 mmol/L])
Serum β-hydroxybutyrate ↑ (0.50 mmol/L, [0.02–0.27 mmol/L])

Serum β-hydroxybutyrate ↑ (0.38 mmol/L, [0.02–0.27 mmol/L])


Serum β-hydroxybutyrate ↑ (0.35 mmol/L, [0.02–0.27 mmol/L])

Serum β-hydroxybutyrate ↑ (0.45 mmol/L, [0.02–0.27 mmol/L])

Serum β-hydroxybutyrate ↑ (0.57 mmol/L, [0.02–0.27 mmol/L])

0.070 h (3–4 min), and the average CL ranged from 5.2 to 7.3 L/h.
Values of t1/2, Tmax, and CL were not significantly affected by increasing
dose. However, Cmax and AUC0–∞increased in a similar ratio over the
full range of doses tested. For dose proportionality analysis of adam-
gammadex (Fig. 2), the estimate of slope was close to 1, and it was not
Urine ketone body ↑ (1.0 (1+) mmol/L, [-])

Urine ketone body ↑ (1.0 (1+) mmol/L, [-])

significantly different from 1 after single doses of 2, 4, 8, 16, 24, and


Urinary glucose ↑ (3.9 (1+) mmol/L, [-])

Urinary glucose ↑ (17 (3+) mmol/L, [-])

32 mg/kg (AUC0–∞, 0.9965 [90%CI, 0.9468, 1.046]; Cmax, 0.9462


Adverse event (value, [normal range])

[90%CI, 0.8800, 1.012]). Therefore, adamgammadex exposure in


plasma increased in a dose-proportional manner.
The cumulative amount of adamgammadex excreted in the urine
increased with dose (Table 6). In different dose groups, the mean cu-
mulative fraction of adamgammadex ranged from approximately
73%–93% (Fig. 1B). Broadly, urinary excretion was a significant ex-
cretory pathway for adamgammadex, and was mostly complete at 8 h
after dosing.
Adamgammadex group

4. Discussion
16 mg/kg
16 mg/kg
24 mg/kg
24 mg/kg
24 mg/kg

Sugammadex was developed by modifying γ-cyclodextrin of the


2 mg/kg
4 mg/kg
4 mg/kg
8 mg/kg
8 mg/kg
8 mg/kg
Table 4

parent structure and adding mercaptopropionic sodium side chains


Dose

(Abad-Gurumeta et al., 2015), which reduce the “molecular binding

4
Y. Jiang, et al. European Journal of Pharmaceutical Sciences 141 (2020) 105134

Table 5
Pharmacokinetic parameters of adamgammadex sodium in plasma.
Parameter Geometric mean (GCV%)
2 mg/kg 4 mg/kg 8 mg/kg 16 mg/kg 24 mg/kg 32 mg/kg

AUC0–∞ (µg*h/mL) 15.24 (10.7) 41.39 (18.5) 77.29 (14.9) 143.2 (10.7) 215.8 (15.0) 251.1 (14.8)
AUC0–t (µg*h/mL) 15.03 (10.7) 40.96 (18.4) 76.42 (14.3) 142.5 (10.9) 215.3 (15.0) 249.8 (14.5)
AUC0–24 (µg*h/mL) 15.42 (11.5) 42.17 (18.7) 78.72 (15.3) 143.6 (10.9) 216.4 (15.1) 251.7 (14.9)
Cmax (µg/mL) 23.92 (16.3) 47.07 (19.3) 120.2 (17.6) 194.5 (29.2) 250.9 (17.7) 334.7 (24.6)
Tmax (h) [a] 0.050 (0.03–0.05) 0.070 (0.05–0.10) 0.050 (0.05–0.07) 0.070 (0.05–0.08) 0.070 (0.05–0.07) 0.070 (0.05–0.08)
t1/2 (h) 1.616 (11.3) 1.644 (4.5) [b] 1.685 (18.1) 1.963 (13.6) 1.755 (8.0) 1.898 (12.2)
CL (L/h) 7.042 (17.3) 5.223 (12.5) 5.647 (9.1) 6.338 (19.2) 6.416 (20.2) 7.298 (12.6)
Vd (L) 16.41 (25.5) 12.36 (17.2) 13.72 (19.2) 17.98 (28.2) 16.23 (18.1) 19.97 (9.6)

GCV% = percent coefficient of variation of geometric mean; AUC = area under the curve; t1/2 = half-time; CL = clearance; Vd = volume of distribution.
[a]
Presented as median (minimum–maximum).
[b]
n = 6. Subject 022 in the 4 mg/kg dose group had missing samples at 2, 3, 4 min. Except for t1/2, the rest of the PK parameters could not be reliably estimated
and therefore were excluded from summary statistics.

specificity” of sugammadex due to the absence of chiral carbons. With a placebo, and there was no AEs specific to adamgammadex. All volun-
large number of such carboxyl groups, sugammadex may also capture teers experiencing AEs without treatment or intervention. In contrast,
non-targeted substances, like intrinsic proteins, in vivo, which may lead sugammadex is associated with side effects such as hypotension, dry
to produce the potential risks of side effects. As mentioned above, su- mouth, nausea, anaphylaxis, bleeding, and prolonged QT interval
gammadex was proven that it increased the risk of allergic reactions (Fuchs-Buder et al., 2013; Gijsenbergh et al., 2005; Takazawa et al.,
and postoperative bleeding (Baldo and McDonnell, 2014; Carron, 2015; 2016). These side effects were not present in adamgammadex.
Rahe-Meyer et al., 2014; Takazawa et al., 2014; Tsur and In preclinical pharmacokinetic studies (Chen et al., 2015; Qi et al.,
Kalansky, 2014). Strengthening the chirality of the cyclodextrin could 2018), no metabolites of adamgammadex have been identified in the
lead to improvement in the molecular binding specificity. Adamgam- hepatocytes from various species, plasma, or urine. The main dis-
madex, developed by Hangzhou Adamerck Pharmlabs Inc., introduced tribution and excretion of adamgammadex in rats occurs in the kidney,
amino acid derivatives with chiral carbons to each sugar unit of γ-cy- where it is excreted without any biotransformation reactions. Measured
clodextrin, enhancing at least 20% of chiral carbon atoms and greatly by the isothermal titration calorimetry, adamgammadex, similar to
improving specificity. As the modified γ-cyclodextrin derivative, sugammadex, combined with rocuronium at an equivalent molar ratio
adamgammadex is composed of a lipophilic core and a hydrophilic for reversing its neuromuscular blockade effect. After a single injection
periphery which forms an inactive and tight complex with free rocur- in healthy subjects, the half-time, clearance, renal clearance rate, ap-
onium or vecuronium molecules. Determined by the isothermal titra- parent volume of distribution, and mean residence time had minimal
tion calorimetry, adamgammadex combined with rocuronium at an changes. Compared with sugammadex (Gijsenbergh et al., 2005;
equivalent molar ratio for reversing its neuromuscular blockade effect Sorgenfrei et al., 2006; Sparr et al., 2007; Staals et al., 2010), adam-
(Chen et al., 2015). Furthermore, the potential side effects induced by gammadex had a lower t1/2 value of 1.8 h and higher urinary excretion
sugammadex such as sensitization, bleeding and HR (including ven- ratio of 83%. With rapid clearance and non-biotransformation reac-
tricular fibrillation, ventricular tachycardia, cardiac arrest, and brady- tions, adamgammadex yielded rapid urinary excretion of combined
cardia) were measured in the zebrafish and beagle dog models rocuronium and fewer potential risks related to drug accumulation.
(Qi et al., 2018). The results from the preclinical pharmacological and Notably, the required adamgammadex dose might be greater than su-
toxicological studies demonstrated that adamgammadex had a similar gammadex to generate equivalent effects. However, adamgammadex at
efficacy and fewer side effects than sugammadex. Therefore, the added 2, 8, and 32 mg/kg performed as well as sugammadex at 8 mg/kg in
chiral carbon atoms in the γ-cyclodextrin increases the steric hindrance reversing the effect of rocuronium in beagle dogs.
of carboxyl groups which reduced the binding rate with non-targeted Adamgammadex is expected to avoid the side effects of su-
molecules to decrease the potential risks of anaphylaxis and other side gammadex and provide an effective and safe option for clinical an-
effects. esthesia. All results from animal experiments and the phase I clinical
In this clinical trial, we found no changes in basic vital signs or trial demonstrate that adamgammadex can be expected to be a safer
clinically meaningful ECG abnormalities, and all AEs were mild. relaxant binding agent.
Further, the frequency of AEs in adamgammadex was similar to that of

Fig. 2. The dose-normalized individual value and geometric mean of AUC0–∞ (A) and Cmax (B) in healthy volunteers receiving single doses of adamgammadex
sodium (2–32 mg/kg).

5
Y. Jiang, et al. European Journal of Pharmaceutical Sciences 141 (2020) 105134

Table 6
Pharmacokinetic parameters in urine (mean ± CV%).
2 4 8 16 24 32
mg/kg mg/kg mg/kg mg/kg mg/kg mg/kg

Ae(0–24) (µg) 90,200 ± 6.2 161,900 ± 39.5 404,300 ± 21.9 864,800 ± 23.3 1,255,000 ± 15.3 1,513,000 ± 18.0
fe(0–24) (%) 86.4 ± 7.5 73.3 ± 29.5 91.2 ± 9.9 93.2 ± 2.6 90.1 ± 5.9 82.1 ± 14.2
CLr (L/h) 5.9 ± 13.9 3.8 ± 35.5[a] 5.2 ± 15.4 5.9 ± 19.5 5.9 ± 20.4 6.1 ± 19.5

CV% = coefficient of variation; AUC = area under the curve; fe = fractional excretion occurs through the kidneys; CLr = renal clearance rate.
[a]
Missing the plasma sample at 2, 3, 4 min (subject 022), AUC0–24 for that subject was not reliably for calculating the CLr.

5. Conclusions Spectroscopy study on the interaction of γ-cyclodextrin and its two derivatives with
rocuronium and vecuronium. Acad. J. Second Mil. Med. Uni. 36, 507–512.
FDA, 2001. Guidance for Industry: Bioanalytical Method Validation. US Department of
In this study, adamgammadex sodium for injection was safe and Health and Human Services, Food and Drug Administration, Center for Drug
well tolerated up to a dose of 32 mg/kg. No drug-related serious AEs Evaluation and Research, Rockville, MD.
were found when administration to 38 healthy subjects, and the overall Fuchs-Buder, T., Meistelman, C., Raft, J., 2013. Sugammadex: clinical development and
practical use. Korean J. Anesthesiol. 65, 495–500.
frequency of AEs in adamgammadex was similar for that in placebo. Gijsenbergh, F., Ramael, S., Houwing, N., van Iersel, T., 2005. First human exposure of
Pharmacokinetic variables remained unchanged regardless of the dose, org 25969, a novel agent to reverse the action of rocuronium bromide.
indicating a linear pharmacokinetic property of adamgammadex over Anesthesiology 103, 695–703.
Groudine, S.B., Soto, R., Lien, C., Drover, D., Roberts, K., 2007. A randomized, dose-
the dose range. Meanwhile, adamgammadex was primarily excreted by finding, phase II study of the selective relaxant binding drug, sugammadex, capable
the kidney. All the results in this study showed that adamgammadex of safely reversing profound rocuronium-induced neuromuscular block. Anesth Analg
may be a novel safe neuromuscular blockade reversal agent. 104, 555–562.
Murphy, G.S., 2018. Neuromuscular monitoring in the perioperative period. Anesth.
Analg. 126, 464–468.
CRediT authorship contribution statement Murphy, G.S., Szokol, J.W., Franklin, M., Marymont, J.H., Avram, M.J., Vender, J.S.,
2004. Postanesthesia care unit recovery times and neuromuscular blocking drugs: a
YingYing Jiang: Data curation, Formal analysis, Writing - original prospective study of orthopedic surgical patients randomized to receive pancuronium
or rocuronium. Anesth. Analg. 98, 193–200 table of contents.
draft. YuJun Zhang: Data curation, Formal analysis, Writing - original Pharmacopoeia, C., 2015. Guidance on Bioanalytical Method Validation.
draft. ShunJu Xiang: Conceptualization. WenLing Zhao: . Jin Liu: Qi, Y., Qi, Y., Yu, B., Lao, Q., Li, C., Zhu, D., Jie, Q., 2018. Screened adamgammadex for
Data curation, Writing - review & editing. WenSheng Zhang: Data reversing rocuronium-induced neuromuscular blockade with greater safety. Chin. J.
Pharmacol. Toxicol. 32, 515–526.
curation, Writing - review & editing. Rahe-Meyer, N., Fennema, H., Schulman, S., Klimscha, W., Przemeck, M., Blobner, M.,
Wulf, H., Speek, M., McCrary Sisk, C., Williams-Herman, D., Woo, T., Szegedi, A.,
Declaration of Competing Interest 2014. Effect of reversal of neuromuscular blockade with sugammadex versus usual
care on bleeding risk in a randomized study of surgical patients. Anesthesiology 121,
969–977.
None. Sacan, O., White, P.F., Tufanogullari, B., Klein, K., 2007. Sugammadex reversal of ro-
curonium-induced neuromuscular blockade: a comparison with neostigmine-glyco-
pyrrolate and edrophonium-atropine. Anesth. Analg. 104, 569–574.
Funding Shields, M., Giovannelli, M., Mirakhur, R.K., Moppett, I., Adams, J., Hermens, Y., 2006.
Org 25969 (sugammadex), a selective relaxant binding agent for antagonism of
This work was supported by the National Major Scientific and prolonged rocuronium-induced neuromuscular block. Br. J. Anaesth. 96, 36–43.
Singh, R., Sadiq, N.M., 2019. Cholinesterase Inhibitors. StatPearls, Treasure IslandFL.
Technological Special Project (grant numbers 2012ZX09103-101-012,
Sorgenfrei, I.F., Norrild, K., Larsen, P.B., Stensballe, J., Ostergaard, D., Prins, M.E., Viby-
2016ZX09101066), and the National Innovation Fund (grant number Mogensen, J., 2006. Reversal of rocuronium-induced neuromuscular block by the
12C26213302758). selective relaxant binding agent sugammadex: a dose-finding and safety study.
Anesthesiology 104, 667–674.
Sparr, H.J., Vermeyen, K.M., Beaufort, A.M., Rietbergen, H., Proost, J.H., Saldien, V.,
Acknowledgments Velik-Salchner, C., Wierda, J.M., 2007. Early reversal of profound rocuronium-in-
duced neuromuscular blockade by sugammadex in a randomized multicenter study:
We are grateful to all experts and staffs for their assistance in this efficacy, safety, and pharmacokinetics. Anesthesiology 106, 935–943.
Staals, L.M., Snoeck, M.M., Driessen, J.J., van Hamersvelt, H.W., Flockton, E.A., van den
clinical study: Dr. Jia Miao from Safety Committee, West China Heuvel, M.W., Hunter, J.M., 2010. Reduced clearance of rocuronium and su-
Hospital, Sichuan University; Dr. Ye Zhu, Dr. Jingyi Li, Dr. Lichuan gammadex in patients with severe to end-stage renal failure: a pharmacokinetic
Yang, et al., Division of Phase I Clinical Studies and Pain Management, study. Br. J. Anaesth. 104, 31–39.
Suy, K., Morias, K., Cammu, G., Hans, P., van Duijnhoven, W.G., Heeringa, M., Demeyer,
West China Hospital, Sichuan University; Dr. Shuning Zhang, Huilan I., 2007. Effective reversal of moderate rocuronium- or vecuronium-induced neuro-
Hu, Jiaqi Lu, Ying Gu, Jiao Wei, Yuan Tian, and Baochun Yu, Adamerck muscular block with sugammadex, a selective relaxant binding agent. Anesthesiology
team members. Special gratitude goes to all subjects in this clinical trial. 106, 283–288.
Takazawa, T., Mitsuhata, H., Mertes, P.M., 2016. Sugammadex and rocuronium-induced
anaphylaxis. J. Anesth. 30, 290–297.
References Takazawa, T., Tomita, Y., Yoshida, N., Tomioka, A., Horiuchi, T., Nagata, C., Orihara, M.,
Yamada, M.H., Saito, S., 2014. Three suspected cases of sugammadex-induced ana-
phylactic shock. BMC Anesthesiol. 14, 92.
Abad-Gurumeta, A., Ripolles-Melchor, J., Casans-Frances, R., Espinosa, A., Martinez-
Tsur, A., Kalansky, A., 2014. Hypersensitivity associated with sugammadex administra-
Hurtado, E., Fernandez-Perez, C., Ramirez, J.M., Lopez-Timoneda, F., Calvo-Vecino,
tion: a systematic review. Anaesthesia 69, 1251–1257.
J.M., Evidence Anaesthesia Review, G., 2015. A systematic review of sugammadex vs
Vanacker, B.F., Vermeyen, K.M., Struys, M.M., Rietbergen, H., Vandermeersch, E.,
neostigmine for reversal of neuromuscular blockade. Anaesthesia 70, 1441–1452.
Saldien, V., Kalmar, A.F., Prins, M.E., 2007. Reversal of rocuronium-induced neu-
Baldo, B.A., McDonnell, N.J., 2014. Sugammadex and anaphylaxis in the operating
romuscular block with the novel drug sugammadex is equally effective under
theater. Rev. Esp. Anestesiol. Reanim. 61, 239–245.
maintenance anesthesia with propofol or sevoflurane. Anesth Analg 104, 563–568.
Brown, S.G., 2004. Clinical features and severity grading of anaphylaxis. J. Aller. Clin.
Viswanathan, C., Bansal, S., Booth, B., DeStefano, A.J., Rose, M.J., Sailstad, J., Shah, V.P.,
Immun. 114, 371–376.
Skelly, J.P., Swann, P.G., Weiner, R., 2007. Quantitative bioanalytical methods va-
Carron, M., 2015. Bleeding risk in surgical patients receiving sugammadex: definitive
lidation and implementation: best practices for chromatographic and ligand binding
conclusions are not yet possible. Anesthesiology 123, 1212.
assays. Pharm. Res. 24, 1962–1973.
Chen, M.-.X., Zhang, W.-.T., Wen, J., Zhao, X., Fan, G.-.R., Qi, Y.-.M., Jie, Q., 2015.

You might also like