ArticlePDF Available

Penehyclidine hydrochloride for treating postoperative nausea and vomiting after laparoscopic bariatric surgery: a double-blinded randomized controlled trial

Authors:

Abstract and Figures

Background Postoperative nausea and vomiting (PONV) is a common and distressing complication of laparoscopic bariatric surgery (LBS). Penehyclidine hydrochloride has been reported to be effective in preventing PONV. Considering the potential preventive effects of penehyclidine against PONV, we hypothesized that intravenous infusion of penehyclidine may alleviate PONV within the first 48 h in patients scheduled for LBS. Methods Patients who underwent LBS were randomly assigned (1:2) to receive saline (Control group, n = 113) or a single intravenous dose of penehyclidine 0.5 mg (PHC group, n = 221). The primary outcome was incidence of PONV within the first 48 h postoperatively. Secondary endpoints included severity of PONV, need for rescue antiemetic therapy, volume of water intake, and time to first flatus. Results PONV occurred in 159 (48%) patients within the first 48 h postoperatively, including 51% in the Control group and 46% in the PHC group. There was no significant difference in the incidence or severity of PONV between the two groups (P > 0.05). Within the first 24 h and 24–48 h, no significant difference was found in incidence or severity of PONV, postoperative nausea, postoperative vomiting, need for rescue antiemetic therapy, or volume of water intake (P > 0.05). Kaplan–Meier curves showed that penehyclidine was significantly associated with a prolonged time to first flatus (median onset time: 22 h vs. 21 h, P = 0.036). Conclusions Penehyclidine did not decrease incidence and severity of PONV in patients undergoing LBS. However, a single intravenous dose of penehyclidine (0.5 mg) was associated with a slightly prolonged time to first flatus. Trial registration Chinese Clinical Trial Registry (ChiCTR2100052418, http://www.chictr.org.cn/showprojen.aspx?proj=134893, date of registration: 25/10/2021).
This content is subject to copyright. Terms and conditions apply.
RESEARCH Open Access
© The Author(s) 2023. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use,
sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and
the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this
article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included
in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will
need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The
Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available
in this article, unless otherwise stated in a credit line to the data.
Ding et al. BMC Anesthesiology (2023) 23:135
https://doi.org/10.1186/s12871-023-02078-0 BMC Anesthesiology
Xiahao Ding and Dapeng Chen contributed equally to this study.
*Correspondence:
Bo Gui
guibosy@163.com
1Department of Anesthesiology and Perioperative Medicine, 1st Affiliated
Hospital with Nanjing Medical University, Nanjing 210029, China
2Department of Anesthesiology, The Huai’an Maternity and Child
Healthcare Hospital, Huai’an 223002, China
3Department of Anesthesiology, Jiangsu Province Official Hospital,
Nanjing 210009, China
4Department of General Surgery, 1st Affiliated Hospital with Nanjing
Medical University, Nanjing 210029, China
Abstract
Background Postoperative nausea and vomiting (PONV) is a common and distressing complication of laparoscopic
bariatric surgery (LBS). Penehyclidine hydrochloride has been reported to be effective in preventing PONV.
Considering the potential preventive effects of penehyclidine against PONV, we hypothesized that intravenous
infusion of penehyclidine may alleviate PONV within the first 48 h in patients scheduled for LBS.
Methods Patients who underwent LBS were randomly assigned (1:2) to receive saline (Control group, n = 113) or a
single intravenous dose of penehyclidine 0.5 mg (PHC group, n = 221). The primary outcome was incidence of PONV
within the first 48 h postoperatively. Secondary endpoints included severity of PONV, need for rescue antiemetic
therapy, volume of water intake, and time to first flatus.
Results PONV occurred in 159 (48%) patients within the first 48 h postoperatively, including 51% in the Control
group and 46% in the PHC group. There was no significant difference in the incidence or severity of PONV between
the two groups (P > 0.05). Within the first 24 h and 24–48 h, no significant difference was found in incidence or
severity of PONV, postoperative nausea, postoperative vomiting, need for rescue antiemetic therapy, or volume of
water intake (P > 0.05). Kaplan–Meier curves showed that penehyclidine was significantly associated with a prolonged
time to first flatus (median onset time: 22 h vs. 21 h, P = 0.036).
Conclusions Penehyclidine did not decrease incidence and severity of PONV in patients undergoing LBS. However, a
single intravenous dose of penehyclidine (0.5 mg) was associated with a slightly prolonged time to first flatus.
Trial registration Chinese Clinical Trial Registry (ChiCTR2100052418, http://www.chictr.org.cn/showprojen.
aspx?proj=134893, date of registration: 25/10/2021).
Keywords Laparoscopic bariatric surgery, Penehyclidine hydrochloride, Postoperative nausea and vomiting, Time to
first flatus
Penehyclidine hydrochloride for treating
postoperative nausea and vomiting after
laparoscopic bariatric surgery: a double-
blinded randomized controlled trial
Xiahao Ding1†, Dapeng Chen1†, Jinxing Che1,2, Siyang Xu1,3, Hui Liang4 and Bo Gui1*
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 2 of 9
Ding et al. BMC Anesthesiology (2023) 23:135
Background
Laparoscopic bariatric surgery (LBS) has been gener-
ally recognized as a safe and effective procedure for
obese adults, given the global prevalence of overweight
and obesity [1, 2]. Postoperative nausea and vomiting
(PONV) is a common and distressing complication after
a surgery involving general anesthesia. PONV occurs
frequently after LBS, at an incidence of 35–89% [3, 4].
Moreover, PONV has been shown to be strongly corre-
lated with several adverse clinical consequences, such as
patient dissatisfaction, incision disruption, water-elec-
trolyte imbalance, prolonged length of hospital stay, and
increased medical expenses [3, 5]. As such, increasing
attention has been paid to this common and burdensome
complaint. Multiple intervention strategies are required
to prevent and manage PONV after LBS.
e etiology of PONV is multifactorial involving
patient, surgical, and anesthesia-related factors. Several
types of neurotransmitters, including serotonin, dopa-
mine, muscarine, neurokinin-1, opioids, and histamine
are closely associated with PONV [6]. Stimulation of ves-
tibular-cochlear, glossopharyngeal, and vagus nerves may
also be responsible for occurrence of PONV. Preclinical
and clinical evidence revealed that multi-modal approach
to target these pharmacological sites is important for
controlling PONV [7]. Penehyclidine hydrochloride is
an anticholinergic drug widely used for the treatment of
organic phosphorus poisoning, as preanesthetic medica-
tion, and for the protection of certain visceral organs [8].
Prophylactic medication with penehyclidine may prevent
PONV in patients undergoing gynecological laparoscopic
surgery, bimaxillary orthognathic surgery, and strabismus
surgery [911]. e underlying mechanism of penehycli-
dine may be related to the inactivation of the M3 mus-
carinic acetylcholine receptor [12, 13]. A combination of
prophylactic antiemetic drugs with different mechanisms
of action should be administered to patients with moder-
ate to high risk of developing PONV.
Considering the potential preventive effects of pene-
hyclidine against PONV, we hypothesized that intrave-
nous infusion of penehyclidine may alleviate PONV and
improve postoperative recovery within the first 48h in
patients scheduled for LBS.
Methods
Study design and ethical approval
is prospective, double-blinded, randomized con-
trolled trial was conducted in an accredited bariatric
center of a tertiary hospital in China between 01/11/2021
and 13/05/2022. e study protocol was approved by
the Ethics Committee (No. 2020-SR-059, 11/03/2020)
and registered in the Chinese Clinical Trial Registry
(ChiCTR2100052418, 25/10/2021). All participants
signed an informed consent form before enrollment in
the study. is study is reported in line with the Con-
solidated Standards of Reporting Trials (CONSORT)
guidelines.
Study participants
e same surgeon performed all procedures, includ-
ing laparoscopic sleeve gastrectomy (LSG), laparoscopic
sleeve gastrectomy plus duodenojejunal bypass (LSG-
DJB), laparoscopic sleeve gastrectomy plus jejunojejunal
bypass (LSG-JJB), and one anastomosis gastric bypass
(OAGB). e inclusion criteria were the American Soci-
ety of Anesthesiologists (ASA) physical status I–III, aged
18–60 years, and scheduled for elective LBS under gen-
eral anesthesia. e exclusion criteria were listed as fol-
lowed: contraindication to penehyclidine administration
(glaucoma, basal ganglia disease, Parkinson’s disease,
pheochromocytoma, myasthenia gravis, severe central
nervous depression, and ECG showing prolonged Q-T
interval) and already using antiemetic drugs within 48h
before surgery.
Randomization and blinding
e participants were randomly assigned (1:2) using a
computer-generated randomization system to receive
saline (Control group) or a single intravenous dose of
penehyclidine 0.5 mg (PHC group) (Chengdu Lisite
Pharmaceutical Co., Ltd., Chengdu, China) after enroll-
ment into the trial. e random allocation sequence was
sealed in opaque envelopes. Participants, care providers,
and investigators were all blinded to treatment allocation.
For safety reasons, the anesthesiologists responsible for
administering the anesthetic were aware of the grouping.
However, they were not responsible for the postoperative
assessment and data collection.
Anesthesia protocol
After the patients arrived in the operating room, elec-
trocardiogram (ECG), blood pressure (BP), oxygen
saturation (SpO2), heart rate (HR), end-tidal carbon
dioxide (EtCO2), and body temperature (T) were moni-
tored. All patients received a loading infusion of lactated
Ringer’s solution (10 ml/kg). Anesthesia induction was
accomplished using dexamethasone 10 mg, midazolam
0.05mg/kg, propofol 1.5‒2.5mg/kg, fentanyl 4–6µg/kg,
plus rocuronium 0.9mg/kg or cis-atracurium 0.15mg/
kg to facilitate tracheal intubation. Meanwhile, patients
in the PHC group received a single intravenous dose of
penehyclidine 0.5mg, while patients in the Control group
received same volume of saline. All patients were endo-
tracheally intubated and mechanically ventilated with
a tidal volume of 6–8 ml/kg (ideal body weight) and a
respiratory rate of 12–16 breaths per minute to main-
tain EtCO2 at the level of 35–45 mmHg. Anesthesia
was maintained with a continuous infusion of propofol
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 3 of 9
Ding et al. BMC Anesthesiology (2023) 23:135
100–200 µg/kg/min, remifentanil 0.05–0.15 µg/kg/min,
rocuronium 5–10µg/kg/min or cis-atracurium 1–3µg/
kg/min. e depth of anesthesia was monitored using
the bispectral index, which was maintained between 40
and 60. Neuromuscular blocking drugs (NMBDs) were
selected according to the choice of reversal agent. Only
patients with a BMI 35kg/m2 undergoing elective LBS
are recommended to receive sugammadex for neuromus-
cular blockade (NMB) reversal in our center. If NMB was
reversed by sugammadex, rocuronium would be used
for both induction and maintenance. If neostigmine, cis-
atracurium would be used instead. e sevoflurane con-
centration was adjusted as necessary. BP was maintained
at a fluctuation of ± 20% of the baseline value by adjusting
the depth of anesthesia or using vasoactive agents during
surgery. All patients received a single intravenous dose of
palonosetron (0.25mg) 30min before the completion of
surgery. Continuous infusion of rocuronium or cis-atra-
curium was stopped after deflation of the pneumoperi-
toneum, and the infusion of propofol and remifentanil
was discontinued at completion of surgery. Patients were
admitted to the post-anesthesia care unit (PACU) with
an endotracheal tube for recovery from anesthesia. We
used clinically physical signs for assessing the recovery of
neuromuscular function and guiding the use of antago-
nists of NMBDs. NMB was allowed to recover sponta-
neously to the moderate block state, and the residual
effects of rocuronium were antagonized via administra-
tion of either sugammadex 200mg or neostigmine 2mg
plus atropine 1mg. Ventilatory support was maintained
until a unified extubation standard was achieved. Tra-
cheal extubation was performed according to a standard-
ized protocol: fully conscious, stable circulation (BP and
HR remain within 20% of the baseline values without any
inotropes), a respiratory rate < 30 breaths/min, maximal
inspiratory pressure < 20 cmH2O, tidal volume > 6 ml/
kg, SpO2 > 93% as well as ability to lift head for 5s. e
criterion used for patient discharge from PACU was the
achievement of a modified Aldrete score 9 [14]. After
patients returned to the ward, an intramuscular injection
of metoclopramide 10 mg was administered as an ini-
tial rescue antiemetic therapy at the request of a patient
or when a patient experienced > 5 episodes of vomiting
within 24 h. If the symptoms persisted one hour after
metoclopramide administration, another 10mg of meto-
clopramide was administered as a second rescue drug.
No more than 20mg of metoclopramide was allowed in
any 24-h period. A total of 10mg of dezocine was admin-
istered intravenously as rescue analgesia in the ward
when the visual analog scale score was 4.
Data collection
Data on clinical characteristics, including age, sex, body
mass index, coexisting disorders, and potential risk
factors for PONV, were collected to assess comparabil-
ity. We also recorded the simplified Apfel score for each
patient [15]. e primary outcome was incidence of
PONV within the first 48 h postoperatively. Secondary
endpoints included severity of PONV, need for rescue
antiemetic therapy, volume of water intake, and time to
first flatus. PONV was defined as at least one episode of
nausea, vomiting, retching, or any combination of these
symptoms. PONV was evaluated as follows: I = no nau-
sea or vomiting, II = nausea but no vomiting, III = mild to
moderate vomiting, and IV = severe and frequent vom-
iting more than five times within 24h. e severity of
postoperative nausea (PON) was assessed with a numeric
rating scale (I = mild, II = moderate, III = severe). e
severity of postoperative vomiting (POV) was recorded
according to the number of vomiting episodes (I = no
vomiting, II = vomiting episodes occurring 1–2 times
within 24h, III = vomiting episodes occurring 3–5 times
within 24 h, IV = vomiting episodes occurring > 5 times
within 24h). e simplified Apfel score contains four risk
factors, including female gender, history of motion sick-
ness or PONV, nonsmoking status, and postoperative
opioid use. Intraoperative opioid consumption means
the total amount of opioids used in the operating room
and PACU. All opioid doses were converted to morphine
intravenous equivalent. According to clinical guidelines,
patients were instructed to drink clear liquids for the first
24–48h after LBS, with the volume gradually increasing
to 2L to promote the recovery of gastrointestinal func-
tion [16]. Accordingly, the volume of postoperative water
intake was measured during the two periods (0–24 h
and 24–48h). e time to first flatus was defined as the
time to the first passage of flatus, as reported by patients,
minus the end time of the surgery.
Sample size
e sample size calculation was based on our preliminary
data, which indicated that 55% of the patients experi-
enced PONV within the first 48h postoperatively. us,
we considered a 35% reduction in the incidence of PONV
as clinically significant. e required sample size was
estimated using Power Analysis and Sample Size software
(PASS, version 15.0. NCSS, LLC. Kaysville, Utah, USA).
e result suggested that 104 patients in the Control
group and 207 in the PHC group would be necessary to
achieve a power of 90% (β = 0.1) with a two-sided confi-
dence interval of 95% (α = 0.05). Considering an attrition
rate of approximately 10%, we increased the sample size
to 346 patients (116 in the Control group and 230 in the
PHC group).
Statistical analysis
Descriptive statistics are presented as mean ± standard
deviation for continuous variables and as frequencies or
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 4 of 9
Ding et al. BMC Anesthesiology (2023) 23:135
proportions for categorical variables. Normal distribu-
tion of the data was confirmed using the Shapiro–Wilk
test. For normally distributed data, an independent Stu-
dent’s t-test was used to assess significant differences
between the two groups. For data with a skewed distri-
bution, the Mann–Whitney U test was used. Categorical
variables were analyzed using either the chi-square test
or Fisher’s exact test, as appropriate. ere were no miss-
ing values in our findings. Statistical significance was set
at P < 0.05. All statistical analyses were performed using
IBM SPSS Statistics for Windows (version 21.0. SPSS Inc.
Chicago, Illinois, USA).
Results
A flow diagram of the study is shown in Fig.1. A total
of 362 patients undergoing elective LBS were assessed for
eligibility, of whom 346 were available for primary analy-
sis between 01/11/2021 and 13/05/2022. Sixteen patients
were excluded before randomization because they did
not meet the inclusion criteria (n = 13), declined to par-
ticipate (n = 1), or case cancellation on the day of surgery
(n = 2). ree patients in the Control group were excluded
because of changes in the surgical procedure during the
operation (n = 2) and unscheduled ICU admission (n = 1).
Nine patients in the PHC group were excluded owing to
temporary alteration of the surgical procedure (n = 5),
severe intraoperative arrhythmia (n = 1), and unsched-
uled ICU admission (n = 3). Ultimately, 113 patients in the
Control group and 221 in the PHC group were examined.
e baseline demographics and intraoperative variables
of patients are shown in Table1. Participant characteris-
tics were similar between the two groups (P > 0.05).
PONV occurred in 159 (48%) patients within the first
48h postoperatively, including 58 (51%) patients in the
Control group and 101 (46%) in the PHC group (Table2).
ere was no significant difference in the incidence or
severity of PONV between the two groups (P > 0.05).
Within the first 24h postoperatively, 48% of the patients
experienced PONV. e incidence of PONV decreased
to 14% in the Control group and 11% in the PHC group
within 24–48 h after surgery (Table 3). As shown in
Table 3; Fig. 2, the incidence of PONV was almost
the same in both groups during the two time periods
(P > 0.05). Specifically, no significant difference was found
in the incidence or severity of PONV, PON, or POV
within the first 48h after LBS (P > 0.05).
As shown in Tables2 and 3, no significant difference
was observed in the proportion of patients receiving
postoperative rescue antiemetic therapy or the amount
Fig. 1 Flow diagram of patients enrolled in the study. ICU intensive care units, PHC penehyclidine hydrochloride
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 5 of 9
Ding et al. BMC Anesthesiology (2023) 23:135
of water intake between the two groups during the two
postoperative periods (P > 0.05).
As shown in Fig.3, the time to first flatus was signifi-
cantly higher in patients in the PHC group (median onset
time, 22h) than in those in the Control group (median
onset time, 21h), as shown by the Kaplan–Meier curves
(P = 0.036).
Discussion
In this prospective, double-blinded, randomized con-
trolled trial, our findings show that 48% of the patients
experienced PONV within the first 48h postoperatively,
with 51% in the Control group and 46% in the PHC
group. Moreover, the expected results were not obtained.
During the early postoperative period, penehyclidine
failed to induce a significant difference with respect to
the incidence and severity of PONV, PON, and POV;
need for rescue antiemetic therapy; or volume of water
intake. However, a single intravenous dose of penehycli-
dine (0.5 mg) was associated with a slightly prolonged
time to first flatus.
e consensus guidelines for the management of
PONV include bariatric surgery as having an increased
risk of PONV [17]. However, the complex etiology and
pathophysiology of PONV remain elusive. e gut vagal
afferent fibers innervate the gastrointestinal tract and
constitute a specific neural pathway to the nucleus of the
solitary tract in the hindbrain that triggers vomiting [18,
19]. Considering that surgical manipulation involves the
gastric vagal nerve, the stimulation of these fibers may
be responsible for the high incidence of PONV after
LBS, especially in LSG. is would explain why patients
undergoing LSG are more prone to develop PONV than
those undergoing other bariatric procedures [3, 4]. More-
over, current evidence strongly suggests a role for the use
of CO2 pneumoperitoneum in the pathogenesis of PONV
[20, 21]. e CO2 pneumoperitoneum is attributed to
increased intra-abdominal pressure, which decreases
intestinal blood flow, especially in obese patients. In
addition to residual intra-abdominal CO2, a recently pub-
lished retrospective study by Lu et al. [22] suggested that
aseptic inflammation caused by ischemia and hypoxia
also plays a role in the occurrence of PONV by inducing
the release of a variety of transmitters. e reason is that
the intestine is probably the most sensitive internal organ
for ischemia. Even short periods of ischemia can induce
the release of neurotransmitters, such as serotonin, that
could stimulate the emetic chemoreceptor trigger zone
Table 1 Baseline demographics and intraoperative variables
Variable Control
group
(n = 113)
PHC group
(n = 221)
P-value
Age (years) 34 ± 9 33 ± 8 0.347
Sex (Male) 25 (22) 68 (31) 0.091
Height (cm) 168 ± 8 168 ± 8 0.909
Weight (kg) 108 ± 25 107 ± 23 0.503
BMI (kg/m2)38 ± 7 38 ± 7 0.391
ASA physical status
II 49 (43) 103 (47) 0.573
III 64 (57) 118 (53)
Apfel risk score
0 2 (2) 12 (5) 0.126
1 13 (11) 39 (18)
2 48 (42) 97 (44)
3 46 (41) 67 (30)
4 4 (4) 6 (3)
Comorbidities
Diabetes mellitus 42 (37) 84 (38) 0.881
Hypertension 22 (20) 43 (20) 0.998
Smoking 17 (15) 46 (21) 0.196
History of motion sickness 3 (3) 6 (3) 0.974
Types of surgery
LSG 70 (62) 134 (61) 0.904
LSG-JJB 28 (25) 55 (25)
LSG-DJB 10 (9) 18 (8)
OAGB 5 (4) 14 (6)
Duration of the anesthesia (min) 93 ± 22 92 ± 23 0.619
Duration of the operation (min) 77 ± 22 76 ± 22 0.867
IOC (mg) 56.8 ± 6.9 58.2 ± 7.5 0.103
Sugammadex 34 (30) 61 (28) 0.635
Neostigmine 79 (70) 160 (72) 0.635
Fluid infusion (ml) 1369 ± 341 1402 ± 370 0.433
Fluid output (ml) 203 ± 67 191 ± 83 0.183
Duration of mechanical ventila-
tion (min)
120 ± 28 120 ± 27 0.768
Duration of PACU stay (min) 66 ± 27 66 ± 23 0.968
Rescue opioids 75 (66) 124 (56) 0.071
Categorical data are presented as n (%), and continuous data are presented as
mean ± standard deviation
Abbreviations: ASA American Society of Anesthesiologists, BMI body mass
index, IOC intraoperative opioids consumption (as intravenous morphine
equivalent), LSG laparoscopic sleeve gastrectomy, LSG-DJB laparoscopic
sleeve gastrectomy plus duodenojejunal bypass, LSG-JJB laparoscopic sleeve
gastrectomy plus jejunojejunal bypass, OAGB one anastomosis gastric bypass,
PACU postanes thesia care unit, PHC penehyclidine hydrochloride
Table 2 Comparison of primary and secondary outcomes within
48 h postoperatively between the two groups
Variable Control
group
(n = 113)
PHC group
(n = 221)
P-
val-
ue
PONV 58 (51) 101 (46) 0.330
PON 12 (11) 22 (10) 0.850
POV 46 (41) 79 (36) 0.377
Rescue antiemetic therapy 30 (27) 49 (22) 0.376
Water intake (ml) 1645 ± 506 1571 ± 515 0.212
Categorical data are presented as n (%), and continuous data are presented as
mean ± standard deviation
Abbreviations: PHC penehyclidine hydrochloride, PONV postoperative nausea
and vomiting, PON postoperative nausea, POV postoperative vomiting
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 6 of 9
Ding et al. BMC Anesthesiology (2023) 23:135
and result in PONV [7, 23]. Given the high levels of
serotonin in the gut, exposing the gut to surgical proce-
dures and anesthetics may increase the excitability of the
gut–vagus–brain reflex that contributes to PONV [24].
Furthermore, PONV is associated with a higher gastric
intraluminal pressure and less distensibility after bariatric
surgery [25, 26].
To date, penehyclidine, as a preanesthetic medica-
tion, is used mainly to reduce respiratory secretions
and inhibit the vagus nerve reflex without increasing
the heart rate. Nevertheless, there is insufficient high-
quality evidence to confirm the protective effect of
penehyclidine against PONV. In a recent prospective
study of 228 pediatric patients undergoing strabismus
surgery, patients receiving penehyclidine (0.01 mg/kg,
maximal dose 0.5 mg, intravenously) after anesthesia
induction had a significantly lower incidence and severity
of PONV within 48h postoperatively [11]. e result of
the marked reduction of PONV by penehyclidine admin-
istration was revealed in the pediatric and non-obese
population, with an intervention performed not on gas-
trointestinal tract but for strabismus. However, Zhang
et al. [9] found that the incidence of POV and need for
antiemetic rescue were both lower in patients receiving
Table 3 Comparison of primary and secondary outcomes within the first 24 h and 24–48 h postoperatively between the two groups
Variable 0–24h after surgery P-value 24–48h after surgery P-value
Control
group
(n = 113)
PHC
group
(n = 221)
Control
group
(n = 113)
PHC
group
(n = 221)
PONV 58 (51) 101 (46) 0.330 16 (14) 24 (11) 0.385
Severity of PONV
I 55 (49) 120 (54) 0.481 97 (86) 197 (89) 0.147
II 12 (11) 22 (10) 11 (10) 14 (6)
III 21 (19) 45 (20) 2 (2) 9 (4)
IV 25 (22) 34 (15) 3 (3) 1 (1)
PON 12 (11) 22 (10) 0.850 11 (10) 14 (6) 0.273
Severity of PON
I 5 (4) 6 (3) 0.051 4 (4) 6 (3) 0.236
II 7 (6) 10 (5) 7 (6) 6 (3)
III 0 (0) 6 (3) 0 (0) 2 (1)
POV 46 (41) 79 (36) 0.377 5 (4) 10 (5) 0.967
Severity of POV
I 67 (59) 142 (64) 0.489 108 (96) 211 (96) 0.133
II 6 (5) 15 (7) 2 (2) 6 (3)
III 15 (13) 30 (14) 0 (0) 3 (1)
IV 25 (22) 34 (15) 3 (3) 1 (1)
Rescue antiemetic therapy 30 (27) 49 (22) 0.376 3 (3) 5 (2) 0.826
Water intake (ml) 450 ± 160 459 ± 158 0.628 1195 ± 380 1112 ± 390 0.065
Categoric al data are presented as n (%), and cont inuous data are presente d as mean ± s tandard deviation
Abbreviations: PHC penehyclidine hydrochloride, PONV postoperative nausea and vomiting, PON postoperative nausea, POV postoperative vomiting
Fig. 2 Stacked bar charts showing the severity of PONV (A), PON (B), and POV (C) within 48 h postoperatively between the two groups. The severity of
PONV was evaluated as follows: I = no nausea or vomiting, II = nausea but no vomiting, III = mild to moderate vomiting, and IV = severe and frequent vom-
iting more than five times within 24 h. The severity of PON was evaluated as follows: I = mild, II = moderate, III = severe. The severity of POV was evaluated
as follows: I = no vomiting, II = vomiting episodes occurring 1–2 times within 24 h, III = vomiting episodes occurring 3–5 times within 24 h, IV = vomiting
episodes occurring > 5 times within 24 h. PONV postoperative nausea and vomiting, PON postoperative nausea, POV postoperative vomiting
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 7 of 9
Ding et al. BMC Anesthesiology (2023) 23:135
tropisetron, a long-acting 5-hydroxytryptamine-3 antag-
onist, than in patients receiving penehyclidine (0.01mg/
kg, maximal dose 1.0mg, intramuscularly) after gyneco-
logical laparoscopic surgery. In contrast, a combination
of tropisetron and penehyclidine was more effective in
preventing PONV than monotherapy with either tropi-
setron or penehyclidine. Considering the mean elimina-
tion half-life of penehyclidine from a pharmacokinetic
perspective, a low-dose bolus (0.5 mg, intravenously)
plus continuous infusion (a dose of 0.25mg added to 100
ml at a rate of 2 ml/h for 48h, intravenously) of pene-
hyclidine was confirmed to be more effective in reducing
PONV after bimaxillary orthognathic surgery [27]. We
speculated that the following reasons may be responsible.
First, penehyclidine may reduce the vagus nerve reflex,
which may inhibit vagal afferent activation to mitigate
PONV. Second, penehyclidine has been confirmed to
relieve gastrointestinal smooth muscle spasms by acting
on the cholinergic receptors in the glands and smooth
muscles of the digestive tract, which is effective in low-
ering gastric intraluminal pressure after surgery. ird,
a previous study suggested that penehyclidine post-con-
ditioning could improve small intestinal mucosal injury
and reduce damage to the barrier function of the small
intestinal mucosa caused by limb ischemia-reperfusion
[28]. Accordingly, we presume that improving intesti-
nal microcirculation might be beneficial in decreasing
PONV. However, penehyclidine was not found to be sig-
nificantly associated with a lower incidence of PONV
in our study. A meta-analysis confirmed that LSG is
associated with an increase in the vagal tone [29]. e
dosage of penehyclidine used in the study may have been
inadequate for the effective inhibition of the gut vagal
afferent activation. Considering that the volume of dis-
tribution of the drug in obese patients may change, the
dosage needs to be adjusted to achieve the desired effect.
erefore, we also need to bring in the assessment of the
pharmacokinetics of penehyclidine when we explored its
effects on PONV. Moreover, gastric denervation remains
difficult to avoid during LBS and may weaken penehycli-
dine’s action in relieving gastrointestinal smooth muscle
spasms. In addition, whether penehyclidine has an effect
on intra-abdominal pressure and the release of neu-
rotransmitters warrants further investigation.
Postoperative water intake volume and time to first
flatus are two important indices for evaluating recov-
ery of gastrointestinal function after surgery. Early oral
hydration after laparoscopic cholecystectomy has been
associated with a lower incidence of PONV in the ward
[30]. is may be owing to two possible reasons. First,
patients exhibiting a low incidence of PONV are more
likely to drink more water. Early oral hydration and
increased water intake reduce postoperative gastroin-
testinal tract dysfunction and accelerate gastric emp-
tying, which is beneficial for lowering PONV. Second,
theoretically, penehyclidine use may be associated with
delayed recovery of intestinal functions postoperatively
owing to its anticholinergic effects on the gastrointesti-
nal tract. Indeed, we found that penehyclidine was sig-
nificantly associated with a prolonged time to the first
flatus. But this difference is just one hour, which may be
not clinically significant. In the present study, patients
on penehyclidine treatment also showed a trend of lower
postoperative water intake volume within 24–48h after
surgery, but the difference was not significant. Although
the use of anticholinergic agents has been verified to be
the main independent risk factor for moderate-to-severe
postoperative thirst [31], this effect appeared to be mod-
est after LBS, especially in the ward.
A few issues concerning our study need to be clarified.
Sugammadex and neostigmine are both commonly used
to antagonize NMBDs-induced neuromuscular paralysis
in the clinical setting. Considering its beneficial effects
on providing fast recovery of neuromuscular function, we
tend to recommend sugammadex for the patients with
BMI 35 kg/m2 as a NMB reversal agent in our center
upon their informed consent. erefore, the differences
in the selection of NMBDs and their reversal agents may
call into the question whether the use of these medica-
tion is associated with PONV. However, we detected
no difference in the use of NMB reversal agent between
the two groups. Furthermore, rocuronium and cis-atra-
curium are both intermediate-acting nondepolarizing
NMBDs, which means the effect of the two drugs are
Fig. 3 Kaplan-Meier curves of the time to first flatus postoperatively be-
tween the two groups. CON control; PHC penehyclidine hydrochloride
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 8 of 9
Ding et al. BMC Anesthesiology (2023) 23:135
comparable. Although the train of four is not routinely
used for monitoring of curarization during surgery in our
center, both groups strictly followed the same indications
for extubation and discharge from PACU. Considering
that our primary outcome is the incidence and severity
of PONV during postoperative stay in the ward, it seems
unlikely that the choice of NMBDs and the degree of
intraoperative curarization would influence PONV long
after administration in the present study.
is study has some limitations. First, we just enrolled
patients undergoing elective LBS, who are aged 18–60
years and ASA physical status I–III. Futhermore, this
was a single-center study with relatively small sample
size, which limits the generalizability of the results. e
results of this trial should be considered with caution.
In the future, large multi-center randomized controlled
prospective studies should be conducted to comprehen-
sively observe the safety and efficacy of penehyclidine in
patients undergoing LBS. Second, further in-depth stud-
ies are needed to determine the appropriate dosage of
penehyclidine to prevent PONV in obese patients, which
needs to bring in the assessment of the volume of distri-
bution. An improved understanding of the pathophysi-
ology of PONV in patients undergoing LBS is needed to
guide future studies.
Conclusions
Penehyclidine did not decrease PONV in patients who
underwent LBS. As “metabolic surgery” is increasingly
recognized by the public, further well-designed random-
ized controlled trials are warranted to validate the results
and provide high-quality evidence for improving anti-
emetic treatment for the prevention and management of
PONV.
Abbreviations
ASA American Society of Anesthesiologists
BMI body mass index
BP blood pressure
CONSORT Consolidated Standards of Reporting Trials
ECG electrocardiogram
EtCO2 end-tidal carbon dioxide
HR heart rate
IOC intraoperative opioids consumption
LBS laparoscopic bariatric surgery
LSG-DJB laparoscopic sleeve gastrectomy plus duodenojejunal bypass
LSG-JJB laparoscopic sleeve gastrectomy plus jejunojejunal bypass
NMB neuromuscular blockade
NMBDs neuromuscular blocking drugs
OAGB one anastomosis gastric bypass
PACU post-anesthesia care unit
PHC penehyclidine hydrochloride
PONV postoperative nausea and vomiting
PON postoperative nausea
POV postoperative vomiting
SpO2 oxygen saturation
Acknowledgements
We thank Dr. Yuting Wang for the assistance of data collection. We thank Prof.
Honggang Yi for his statistical analysis assistance. We also would like to thank
Editage (www.editage.cn) for English language editing.
Author Contribution
Xiahao Ding: Data collection, methodology, formal analysis, writing – original
draft; Dapeng Chen: Methodology, formal analysis, writing – original
draft; Jinxing Che: Methodology, formal analysis; Siyang Xu: Methodology,
validation; Hui Liang: Conceptualization; Bo Gui: Conceptualization, formal
analysis, supervision, validation, writing – review and editing. All authors read
and approved the final manuscript.
Funding
This research did not receive any specific grant from funding agencies in the
public, commercial, or not-for-profit sectors.
Data Availability
The datasets generated and/or analyzed during the current study are not
publicly available due to potential patient privacy compromise but are
available from the corresponding author on reasonable request.
Declarations
Ethics approval and consent to participate
This study was approved by the Institutional Review Board of the 1st
Affiliated Hospital with Nanjing Medical University (IRB #2020-SR-059) on
March 11, 2020 and written informed consent was obtained from all subjects
participating in the trial. All methods were performed following the relevant
guidelines and regulations.
Consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
Received: 1 February 2023 / Accepted: 4 April 2023
References
1. Angrisani L, Santonicola A, Iovino P, Ramos A, Shikora S, Kow L. Bariatric
surgery survey 2018: similarities and disparities among the 5 IFSO chapters.
Obes Surg. 2021;31(5):1937–48.
2. Howard R, Chao GF, Yang J, Thumma J, Chhabra K, Arterburn DE, et al. Com-
parative safety of sleeve gastrectomy and gastric bypass up to 5 years after
surgery in patients with severe obesity. JAMA Surg. 2021;156(12):1160–9.
3. Suh S, Helm M, Kindel TL, Goldblatt MI, Gould JC, Higgins RM. The impact
of nausea on post-operative outcomes in bariatric surgery patients. Surg
Endosc. 2020;34(7):3085–91.
4. Zhu J, Wu L, Chen G, Zhao X, Chen W, Dong Z, et al. Preoperative reflux or
regurgitation symptoms are independent predictors of postoperative nausea
and vomiting (PONV) in patients undergoing bariatric surgery : a propensity
score matching analysis. Obes Surg. 2022;32(3):819–28.
5. Kassir R, Debs T, Blanc P, Gugenheim J, Ben Amor I, Boutet C, et al. Complica-
tions of bariatric surgery: presentation and emergency management. Int J
Surg. 2016;27:77–81.
6. Nielsen M, Olsen NV. Genetic polymorphisms in the cytochrome P450 system
and efficacy of 5-hydroxytryptamine type 3 receptor antagonists for postop-
erative nausea and vomiting. Br J Anaesth. 2008;101(4):441–5.
7. Horn CC, Wallisch WJ, Homanics GE, Williams JP. Pathophysiological and
neurochemical mechanisms of postoperative nausea and vomiting. Eur J
Pharmacol. 2014;722:55–66.
8. Wang Y, Gao Y, Ma J. Pleiotropic effects and pharmacological properties of
penehyclidine hydrochloride. Drug Des Devel Ther. 2018;12:3289–99.
9. Zhang Z, Zhuang Y, Ouyang F, Zhang A, Zeng B, Gu M. Penehyclidine
enhances the efficacy of tropisetron in prevention of PONV following gyne-
cological laparoscopic surgery. J Anesth. 2012;26(6):864–9.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 9 of 9
Ding et al. BMC Anesthesiology (2023) 23:135
10. Wang LK , Cheng T, Yang XD, Xiong GL, Li N, Wang DX. Penehyclidine for
prevention of postoperative nausea and vomiting following bimaxillary
orthognathic surgery: a randomized, double-blind, controlled trial. J Anesth.
2022;36(1):122–36.
11. Sun J, Cao X, Lu T, Li N, Min X, Ding Z. Penehyclidine mitigates postopera-
tive nausea and vomiting and intraoperative oculocardiac reflex in patients
undergoing strabismus surgery: a prospective, randomized, double-blind
comparison. BMC Anesthesiol. 2021;21(1):49.
12. Janicki PK, Vealey R, Liu J, Escajeda J, Postula M, Welker K. Genome-wide
Association study using pooled DNA to identify candidate markers mediat-
ing susceptibility to postoperative nausea and vomiting. Anesthesiology.
2011;115(1):54–64.
13. Klenke S, de Vries GJ, Schiefer L, Seyffert N, Bachmann HS, Peters J, et al.
CHRM3 rs2165870 polymorphism is independently associated with post-
operative nausea and vomiting, but combined prophylaxis is effective. Br J
Anaesth. 2018;121(1):58–65.
14. Aldrete JA, Kroulik D. A postanesthetic recovery score. Anesth Analg.
1970;49(6):924–34.
15. Apfel CC, Läärä E, Koivuranta M, Greim CA, Roewer N. A simplified risk score
for predicting postoperative nausea and vomiting: conclusions from cross-
validations between two centers. Anesthesiology. 1999;91(3):693–700.
16. Stenberg E, Dos Reis Falcão LF, O’Kane M, Liem R, Pournaras DJ, Salminen P, et
al. Guidelines for perioperative care in bariatric surgery: enhanced recovery
after surgery (ERAS) society recommendations: a 2021 update. World J Surg.
2022;46(4):729–51.
17. Gan TJ, Belani KG, Bergese S, Chung F, Diemunsch P, Habib AS, et al. Fourth
consensus guidelines for the management of postoperative nausea and
vomiting. Anesth Analg. 2020;131(2):411–48.
18. Gupta RG, Schafer C, Ramaroson Y, Sciullo MG, Horn CC. Role of the abdomi-
nal vagus and hindbrain in inhalational anesthesia-induced vomiting. Auton
Neurosci. 2017;202:114–21.
19. Horn CC. The medical implications of gastrointestinal vagal afferent pathways
in nausea and vomiting. Curr Pharm Des. 2014;20(16):2703–12.
20. Besir A, Tugcugil E. Comparison of different end-tidal carbon dioxide levels
in preventing postoperative nausea and vomiting in gynaecological patients
undergoing laparoscopic surgery. J Obstet Gynaecol. 2021;41(5):755–62.
21. Gin E, Lowen D, Tacey M, Hodgson R. Reduced laparoscopic intra-abdominal
pressure during laparoscopic cholecystectomy and its effect on post-oper-
ative pain: a double-blinded randomised control trial. J Gastrointest Surg.
2021;25(11):2806–13.
22. Lu H, Zheng C, Liang B, Xiong B. Mechanism and risk factors of nausea and
vomiting after TACE: a retrospective analysis. BMC Cancer. 2021;21(1):513.
23. Beuk RJ, Heineman E, Tangelder GJ, Kurvers HA, Bonke HJ, oude Egbrink MG.
Effects of different durations of total warm ischemia of the gut on rat mesen-
teric microcirculation. J Surg Res. 1997;73(1):14–23.
24. Syn NL, Cummings DE, Wang LZ, Lin DJ, Zhao JJ, Loh M, et al. Associa-
tion of metabolic-bariatric surgery with long-term survival in adults with
and without diabetes: a one-stage meta-analysis of matched cohort
and prospective controlled studies with 174 772 participants. Lancet.
2021;397(10287):1830–41.
25. Yehoshua RT, Eidelman LA, Stein M, Fichman S, Mazor A, Chen J, et al. Lapa-
roscopic sleeve gastrectomy–volume and pressure assessment. Obes Surg.
2008;18(9):1083–8.
26. Fathy M, Abdel-Razik MA, Elshobaky A, Emile SH, El-Rahmawy G, Farid A, et
al. Impact of pyloric injection of magnesium sulfate-lidocaine mixture on
postoperative nausea and vomiting after laparoscopic sleeve gastrectomy: a
randomized-controlled trial. Obes Surg. 2019;29(5):1614–23.
27. Ma W, Qi Y, Liu C, Wang M, Zhang Y, Yao W. Effect of individualized treatment
strategy on postoperative nausea and vomiting in gynaecological laparo-
scopic surgery: a double-blind, randomized, controlled trial. BMC Anesthesiol.
2022;22(1):266.
28. Zhang Y, Leng YF, Xue X, Zhang Y, Wang T, Kang YQ. Effects of penehyclidine
hydrochloride in small intestinal damage caused by limb ischemia-reperfu-
sion. World J Gastroenterol. 2011;17(2):254–9.
29. Geronikolou SA, Albanopoulos K, Chrousos G, Cokkinos D. Evaluating the
homeostasis assessment model insulin resistance and the cardiac autonomic
system in bariatric surgery patients: a meta-analysis. Adv Exp Med Biol.
2017;988:249–59.
30. Wu M, Yang L, Zeng X, Wang T, Jia A, Zuo Y, et al. Safety and feasibility of early
oral hydration in the postanesthesia care unit after laparoscopic cholecys-
tectomy: a prospective, randomized, and controlled study. J Perianesth Nurs.
2019;34(2):425–30.
31. Lee CW, Liu ST, Cheng YJ, Chiu CT, Hsu YF, Chao A. Prevalence, risk factors, and
optimized management of moderate-to-severe thirst in the post-anesthesia
care unit. Sci Rep. 2020;10(1):16183.
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional affiliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... 13,14 Previous studies have shown that the mechanism of penehyclidine reducing the incidence of PONV is complex, which may be related to its blocking of the muscarinic acetylcholine receptor, which is involved in the development of PONV through various mechanisms. Recent clinical studies have shown that penehyclidine for prevention of PONV has made good progress in some surgical procedures, such as ophthalmology, 15 laparoscopic bariatric surgery, 16 thyroid surgery 17 and stomatology surgery. 18 Patients undergoing gynecological laparoscopic surgery are at high risk for PONV. ...
... 15 However, Ding et al found that penehyclidine (bolus 5mg, IV) did not reduce the incidence and severity of postoperative PONV in bariatric surgery patients. 16 In a study of patients undergoing gynecological laparoscopic surgery, Zhang et al observed that a single dose of penehyclidine before anesthesia also did not reduce the incidence of PONV. 27 Some of the above studies are based on a single dose of intravenous penehyclidine to investigate the prevention of PONV. ...
Article
Full-text available
Purpose We designed this study to investigate the effect of intravenous use of penehyclidine on postoperative nausea and vomiting (PONV) after gynecological laparoscopic surgery. Patients and Methods Ninety-two Women Patients (Aged ≥ 18) Scheduled for Elective Gynecologic Laparoscopy Were Enrolled in the Current Study. Patients Were Equally Randomized Assigned Into Penehyclidine group (PHC group: received a bolus of penehyclidine 10 μg/kg during the induction of anesthesia, then followed by a continuous infusion of 10 μg/kg penehyclidine at a fixed rate of 2.0 mL/h in postoperative intravenous analgesia pump over 48h, 0.5 mg upper limit respectively) or Control group (received 0.9% saline in replace of penehyclidine at the same time points). The primary outcome measure was the incidence of postoperative nausea and vomiting in the postanesthesia care unit and ward area. Quality of Recovery-15 (QoR-15) scores and general comfort questionnaire (GCQ) scores were assessed on postoperative day (POD) 1, 2. Results Patients between two groups had comparable baseline characteristics. Compared with the Control group, the incidence and severity of PONV, postoperative nausea (PON), and postoperative vomiting (POV) were significantly lower in the PHC group at 2h (PONV: P = 0.002, P = 0.004, respectively; PON: P = 0.018, P = 0.038, respectively; POV: P = 0.011, P = 0.072, respectively), 24h (PONV: P = 0.003, P = 0.001, respectively; PON: P = 0.010, P = 0.032, respectively; POV: P = 0.006, P = 0.044, respectively), and 48h (PONV: P = 0.003, P = 0.002, respectively; PON: P = 0.007, P = 0.019, respectively; POV: P = 0.002, P = 0.013, respectively) after surgery. The QoR-15 and GCQ scores of the PHC group were significantly higher than those of the Control group at POD 1, 2 (P < 0.001; P < 0.001, respectively). Conclusion Our findings suggest that perioperative intravenous application of penehyclidine can effectively prevent postoperative nausea and vomiting in gynecological laparoscopic surgery patients and improve postoperative recovery.
... In this study, we investigated the predictive factors for PONV based on our recently completed trial [13]. We developed and validated a nomogram to predict PONV in patients undergoing LBS. ...
... In the present study, data were retrieved from the database of our recently completed trial (PHCBS), which was a prospective, randomized, double-blind, controlled study performed at an accredited bariatric center of a tertiary hospital in China between November 1, 2021, and May 13, 2022 [13]. This trial was designed to explore the effect of penehyclidine hydrochloride, an anticholinergic drug, on PONV in adult patients scheduled for LBS. ...
Article
Full-text available
Background Postoperative nausea and vomiting (PONV) is a common and distressing complication of laparoscopic bariatric surgery (LBS). However, there is a lack of effective integrated prediction models for preventing and treating PONV in patients after LBS. Methods Based on a randomized controlled trial conducted between November 1, 2021, and May 13, 2022, we included 334 participants who underwent LBS according to the inclusion criteria. The database was divided randomly into training and validation cohorts in a ratio of 7:3. Least absolute shrinkage and selection operator plus multivariable logistic regression were used to identify independent predictors and construct a nomogram. The performance of the nomogram was assessed and validated by the area under the receiver operating characteristic curve (AUC), the concordance index (C-index), calibration plots, and a decision curve analysis (DCA). We also explored specific risk factors for PONV in patients with diabetes. Results The subjects were divided randomly into training (n = 234) and validation (n = 100) cohorts. Age, history of diabetes, type of surgery, and sugammadex use were incorporated to construct a nomogram prediction model. In the training cohort, the AUC and the optimism-corrected C-index were 0.850 [95% confidence interval (CI) 0.801–0.899] and 0.848, while in the validation cohort they were 0.847 (95% CI 0.768–0.925) and 0.844, respectively. The calibration plots showed good agreement between the predicted and actual observations. The DCA results demonstrated that the nomogram was clinically useful. The type of surgery, sugammadex use, and insulin level at 120 min were predictors of PONV in patients with diabetes with an AUC of 0.802 (95% CI 0.705–0.898). Conclusions We developed and validated a prediction model for PONV in patients after LBS. A risk factor analysis of PONV in patients with diabetes provides clinicians with a more precise prophylactic protocol. Graphical abstract
... The combination of serotonin antagonists with dexamethasone is a common dual therapy used to prevent PONV for high-risk patients undergoing various surgical procedures, such as laparoscopic bariatric surgery (palonosetron 0.25 mg plus dexamethasone 10 mg), laparoscopic cholecystectomy (palonosetron 0.075 mg plus dexamethasone 8 mg), and cesarean delivery (palonosetron 0.075 mg plus dexamethasone 4 mg). [10][11][12][13][14] Even with double prophylaxis, PONV may still occur frequently. 5,15 Searching for factors related to PONV in various clinical situations may contribute to better PONV control. ...
Article
Full-text available
Purpose Postoperative nausea and vomiting (PONV) is a major problem after surgery. This study aimed to demonstrate the incidence of PONV and the potential associated factors in female patients undergoing laparoscopic gastrointestinal surgery against the background of double prophylactic therapy. Methods Our retrospective study recruited 109 female patients undergoing laparoscopic gastrointestinal surgery with double prophylactic therapy, combining palonosetron with dexamethasone, from October 2020 to March 2021, at the Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China. Patient characteristics and perioperative management factors were included in univariate and multivariate analyses to identify factors influencing PONV. Results Four patients lacked complete records, and of the 105 patients included in the final analysis, 53 (50.5%) patients developed PONV. Two influencing factors for PONV were identified: a history of chemotherapy (odds ratio [OR] 0.325, 95% confidence interval [CI] 0.123–0.856; p = 0.023) and dosage of hydromorphone ≥ 0.02 mg/kg (OR 2.857, 95% CI 1.247–6.550; p = 0.013). The performance of the multivariate logistic regression was evaluated by analyzing receiver operating characteristic curves, resulting in an area under the curve value of 0.673. Conclusion The incidence of PONV remains high in female patients undergoing laparoscopic gastrointestinal surgery, even with double prophylactic therapy. A dosage of hydromorphone ≥ 0.02 mg/kg may increase risk of PONV, whereas a history of chemotherapy might be a protective factor.
Article
Full-text available
Background: Postoperative nausea and vomiting (PONV) have always been a concern of clinicians and may increase medical costs for patients. Consensus guidelines recommend using multiple antiemetics with different mechanisms as prophylaxis in patients at high risk of PONV. Individualized risk scores for nausea and vomiting and individualized treatment strategies are feasible. This study evaluated the effect of individualized treatment strategies on postoperative nausea and vomiting after laparoscopic gynaecological operations. Methods: This was a double-blind, randomized, controlled trial. A total of 119 adult patients who underwent gynaecological laparoscopic surgery under general anaesthesia were randomly divided into an individualized treatment group or a control group, with the individualized treatment group receiving individualized prevention according to a preoperative risk score of nausea and vomiting and the control group receiving no individualized prevention. Vomiting, retching, nausea, and use of rescue medication were all recorded for 24 h after the operation. The primary outcome variable was complete response, defined as no emesis or the use of rescue medication 24 h postoperatively. Results: The complete response rate was higher in the individualized treatment group (56.7%) than in the control group (23.7%) (95% CI, 0.01-0.27; P < 0.001). The incidences of emesis (18.3% vs. 44.1%, P = 0.002) were significantly lower in the individualized treatment group than in the control group. There were no differences in any nausea (26.7% vs. 33.9%, P = 0.391) or rescue medication use (6.7% vs. 8.5%, P = 0.743). Adverse events and laboratory and electrocardiogram abnormalities occurred no more frequently in the individualized treatment group than in the control group. Conclusion: In conclusion, this single-centre, double-blind, randomized study suggests that an individualized PONV prophylactic treatment strategy based on the number of PONV risk factors could be a safe and effective regimen to reduce the incidence of PONV in adult patients undergoing laparoscopic gynaecological surgery.
Article
Full-text available
Background Postoperative nausea and vomiting (PONV) is common side effect after gastrointestinal surgery. It causses discomfort, increase risk of incision disruption, bleeding and airway blockage. This study aimed to investigate the incidence and severity of PONV and determine whether preoperative reflux or regurgitation symptoms influence PONV in patients undergoing bariatric surgery. Methods Patients with obesity underwent laparoscopic sleeve gastrectomy (LSG) and laparoscopic Roux-en-Y gastric bypass (LRYGB) in our center between January 2016 and December 2018 were divided into PONV and NoPONV group and analyzed retrospectively. PONV and postoperative pain visual analogue scale (VAS) were analyzed after surgery. A 1:1 propensity score matching (PSM) method was performed, and multivariable logistic regression analyses were used to identify predictors for PONV. Results There were 483 patients enrolled, with a mean age of 30.19 ± 9.96 years, and 134 pairs of matched patients were selected from PONV group and NoPONV group after PSM. PONV occurred in 288 (59.6%) patients after bariatric surgery (77.4% after LSG vs 21.5% after LRYGB, p < 0.001). In PONV group, 210 (72.9%) were female, preoperative reflux or regurgitation symptoms were 128 (33.6%). Multivariable analysis found that preoperative reflux or regurgitation symptoms were independent risk factors for PONV after LSG, with an OR of 2.78 (95% CI: 1.12–6.93, p = 0.028). Conclusions Incidence of PONV after bariatric surgery is high. For the first time, this study valued preoperative reflux or regurgitation symptoms as risk factors that may promote PONV after bariatric surgery. Patients with preoperative symptoms undergoing LSG have a higher risk of PONV, so these patients should be carefully evaluated for the feasibility of LSG before surgery.
Article
Full-text available
Background This is the second updated Enhanced Recovery After Surgery (ERAS®) Society guideline, presenting a consensus for optimal perioperative care in bariatric surgery and providing recommendations for each ERAS item within the ERAS® protocol. Methods A principal literature search was performed utilizing the Pubmed, EMBASE, Cochrane databases and ClinicalTrials.gov through December 2020, with particular attention paid to meta-analyses, randomized controlled trials and large prospective cohort studies. Selected studies were examined, reviewed and graded according to the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) system. After critical appraisal of these studies, the group of authors reached consensus regarding recommendations. Results The quality of evidence for many ERAS interventions remains relatively low in a bariatric setting and evidence-based practices may need to be extrapolated from other surgeries. Conclusion A comprehensive, updated evidence-based consensus was reached and is presented in this review by the ERAS® Society.
Article
Full-text available
Purpose To investigate the efficacy and safety of low-dose bolus plus continuous infusion of penehyclidine in preventing postoperative nausea and vomiting (PONV) following bimaxillary surgery. Methods Three hundred fifty-four patients were randomly allocated into three groups. In the Control group, placebo (normal saline) was injected before anesthesia and infused over 48 h after surgery; in the Bolus group, 0.5 mg penehyclidine was injected before anesthesia, whereas placebo was infused after surgery; in the Infusion group, 0.25 mg penehyclidine were injected before anesthesia, another 0.25 mg penehyclidine was infused after surgery. The primary endpoint was the incidence of PONV within 72 h. Results A total of 353 patients were included in intention-to-treat analysis. The PONV incidence was 61.0% (72/118) in the Control group, 40.2% (47/117) in the Bolus group, and 28.0% (33/118) in the Infusion group. The incidence was significantly lower in the Bolus group than in the Control group (RR 0.66; 95% CI 0.51–0.86; adjusted P = 0.003) and in the Infusion group than in the Control group (RR 0.46; 95% CI 0.33–0.63; adjusted P < 0.001); the difference between the Infusion and Bolus groups was not statistically significant (RR 0.70; 95% CI 0.48–1.00; adjusted P = 0.144). Emergence agitation occurred more frequently in the Bolus group than in the Control group (36.8% [43/117] vs. 21.2% [25/118], adjusted P = 0.027), but did not differ significantly between the Infusion and Control groups. Conclusions A low-dose bolus plus continuous infusion of penehyclidine was effective in preventing PONV without increasing emergence agitation. Trial registration Clinicaltrials.gov. Identifier: NCT04454866.
Article
Full-text available
Purpose The mechanism of postoperative nausea and vomiting after TACE is not clear. This study retrospectively analyzed the patient data to explore the mechanism and risk factors of postoperative nausea and vomiting after TACE. Materials and methods The data of 221 patients who underwent TACE in the interventional department from January 2019 to December 2020 were collected. Including: gender, age, liver function before TACE, etiology of liver cirrhosis, BCLC stage of hepatocellular carcinoma, preoperative use of analgesic drugs, preoperative limosis, previous history of vomiting, history of kinetosis, smoking history, history of drinking, chemotherapeutic drugs used during TACE, Dosage of lipiodol, and occurrence of postoperative nausea and vomiting. Results There were 116 cases of nausea after TACE, using binary logistic regression analysis, Sig: ALT0.003; ALP0.000; history of vomiting 0.043; kinetosis 0.006; history of alcohol consumption 0.011; preoperative limosis 0.006; dosage of lipiodol (5–10 mL) 0.029, dosage of lipiodol (> 10 mL) 0.001.There were 89 cases of vomiting after TACE, all accompanied by nausea, Sig: ALP0.000; BCLC stage (B) 0.007; kinetosis 0.034; chemotherapeutic drugs 0.015; dosage of lipiodol (5–10 ml) 0.015, dosage of lipiodol (> 10 ml) 0.000; patients used analgesics before TACE 0.034. Conclusions Causes of post-TACE nausea and vomiting included operative trauma, aseptic inflammation caused by ischemia and hypoxia, chemotherapeutic drugs, ischemia of liver and bile duct, stress and pain during TACE, and patient factors. ALP, BCLC stage, kinetosis, chemotherapeutic drugs, dosage of lipiodol, and preoperative usage of analgesics were risk factors affecting nausea and vomiting after TACE.
Article
Full-text available
Background Metabolic–bariatric surgery delivers substantial weight loss and can induce remission or improvement of obesity-related risks and complications. However, more robust estimates of its effect on long-term mortality and life expectancy—especially stratified by pre-existing diabetes status—are needed to guide policy and facilitate patient counselling. We compared long-term survival outcomes of severely obese patients who received metabolic–bariatric surgery versus usual care. Methods We did a prespecified one-stage meta-analysis using patient-level survival data reconstructed from prospective controlled trials and high-quality matched cohort studies. We searched PubMed, Scopus, and MEDLINE (via Ovid) for randomised trials, prospective controlled studies, and matched cohort studies comparing all-cause mortality after metabolic–bariatric surgery versus non-surgical management of obesity published between inception and Feb 3, 2021. We also searched grey literature by reviewing bibliographies of included studies as well as review articles. Shared-frailty (ie, random-effects) and stratified Cox models were fitted to compare all-cause mortality of adults with obesity who underwent metabolic–bariatric surgery compared with matched controls who received usual care, taking into account clustering of participants at the study level. We also computed numbers needed to treat, and extrapolated life expectancy using Gompertz proportional-hazards modelling. The study protocol is prospectively registered on PROSPERO, number CRD42020218472. Findings Among 1470 articles identified, 16 matched cohort studies and one prospective controlled trial were included in the analysis. 7712 deaths occurred during 1·2 million patient-years. In the overall population consisting 174 772 participants, metabolic–bariatric surgery was associated with a reduction in hazard rate of death of 49·2% (95% CI 46·3–51·9, p<0·0001) and median life expectancy was 6·1 years (95% CI 5·2–6·9) longer than usual care. In subgroup analyses, both individuals with (hazard ratio 0·409, 95% CI 0·370–0·453, p<0·0001) or without (0·704, 0·588–0·843, p<0·0001) baseline diabetes who underwent metabolic–bariatric surgery had lower rates of all-cause mortality, but the treatment effect was considerably greater for those with diabetes (between-subgroup I² 95·7%, p<0·0001). Median life expectancy was 9·3 years (95% CI 7·1–11·8) longer for patients with diabetes in the surgery group than the non-surgical group, whereas the life expectancy gain was 5·1 years (2·0–9·3) for patients without diabetes. The numbers needed to treat to prevent one additional death over a 10-year time frame were 8·4 (95% CI 7·8–9·1) for adults with diabetes and 29·8 (21·2–56·8) for those without diabetes. Treatment effects did not appear to differ between gastric bypass, banding, and sleeve gastrectomy (I² 3·4%, p=0·36). By leveraging the results of this meta-analysis and other published data, we estimated that every 1·0% increase in metabolic–bariatric surgery utilisation rates among the global pool of metabolic–bariatric candidates with and without diabetes could yield 5·1 million and 6·6 million potential life-years, respectively. Interpretation Among adults with obesity, metabolic–bariatric surgery is associated with substantially lower all-cause mortality rates and longer life expectancy than usual obesity management. Survival benefits are much more pronounced for people with pre-existing diabetes than those without. Funding None.
Article
Full-text available
Background Postoperative nausea and vomiting (PONV) is one of the most frequent complications following strabismus surgery. Penehyclidine, an anticholinergic agent, is widely used as premedication. This study investigated the effect of preoperative penehyclidine on PONV in patients undergoing strabismus surgery. Methods In this prospective, randomized, double-blind study, patients scheduled for strabismus surgery under general anesthesia were randomly assigned to either penehyclidine ( n = 114) or normal saline ( n = 104) group. Penehyclidine was administrated immediately after anesthesia induction, and normal saline was substituted as control. PONV was investigated from 0 to 48 h after surgery. Intraoperative oculocardiac reflex (OCR) was also recorded. Results Compared with normal saline, penehyclidine significantly reduced PONV incidence (30.7% vs. 54.8%, P < 0.01) and mitigated PONV severity as indicated by severity scoring ( P < 0.01). Compared with normal saline, penehyclidine also significantly reduced OCR incidence (57.9% vs. 77.9%, P < 0.01) and mitigated OCR severity, as indicated by the requirement for atropine rescue (77.3% vs. 90.1%, P < 0.05) and the maximum decrease of heart rate during OCR (23.1 ± 9.4 bpm vs. 27.3 ± 12.4 bpm, P < 0.05). The recovery course did not differ between groups. Conclusions Penehyclidine administrated after anesthesia induction significantly reduced the incidence of PONV and alleviated intraoperative OCR in patients undergoing strabismus surgery. Trial registration ClinicalTrials.gov ( NCT04054479 ). Retrospectively registered August 13, 2019.
Article
Full-text available
Background The 2018 IFSO Survey focused on similarities and disparities in the number and types of surgical and endoluminal interventions among the IFSO chapters occurred in the last decade. Methods All IFSO Societies were asked to fill in the IFSO survey form on how many and which surgical and endoluminal interventions have been performed in the 2018. A special section was added, asking about the existence of national guidelines for bariatric and metabolic surgery, national recommendations for preoperative gastroscopy, type of reimbursement for bariatric/metabolic surgery, including for patients with BMI < 35 kg/m². The trend analyses from 2008 to 2018 were also performed. Results Fifty-seven/65 (87.7%) IFSO Societies submitted completed forms. The American Society for Metabolic and Bariatric Surgery did not fill the IFSO survey form but provided an official report on the performed interventions. The total number of surgical and endoluminal procedures performed in 2018 in the world was 696,191. Sleeve gastrectomy (SG) remained the most commonly performed bariatric procedure (N = 386,096; 55.4%). Among the total reported bariatric/metabolic interventions, 604,223 (86.8%) were primary surgical and 29,167 (4.2%) primary endoluminal operations; 62,801 (9%) were revisional procedures. Conclusions There was only a slight increase (10.317 procedures) in the total number of the reported bariatric interventions compared to the last IFSO survey. SG continues to be the most commonly performed operation since 2014. The one anastomosis gastric bypass (OAGB), currently the third most performed procedure after SG and Roux-en-Y gastric bypass (RYGB), continues to rise in popularity worldwide.
Article
Importance: Sleeve gastrectomy is the most widely used bariatric operation; however, its long-term safety is largely unknown. Objective: To compare the risk of mortality, complications, reintervention, and health care use 5 years after sleeve gastrectomy and gastric bypass. Design, setting, and participants: This retrospective cohort study included adult patients in a national Medicare claims database who underwent sleeve gastrectomy or gastric bypass from January 1, 2012, to December 31, 2018. Instrumental variables survival analysis was used to estimate the cumulative incidence of outcomes up to 5 years after surgery. Exposures: Laparoscopic sleeve gastrectomy and laparoscopic Roux-en-Y gastric bypass. Main outcomes and measures: The main outcome was risk of mortality, complications, and reinterventions up to 5 years after surgery. Secondary outcomes were health care use after surgery, including hospitalization, emergency department (ED) use, and total spending. Results: Of 95 405 patients undergoing bariatric surgery, 57 003 (60%) underwent sleeve gastrectomy (mean [SD] age, 57.1 [11.8] years), of whom 42 299 (74.2%) were women; 124 (0.2%) were Asian; 10 101 (17.7%), Black; 1951 (3.4%), Hispanic; 314 (0.6%), North American Native; 43 194 (75.8%), White; 534 (0.9%), of other race or ethnicity; and 785 (1.4%), of unknown race or ethnicity. A total of 38 402 patients (40%) underwent gastric bypass (mean [SD] age, 55.9 [11.7] years), of whom 29 050 (75.7%) were women; 109 (0.3%), Asian; 6038 (15.7%), Black; 1215 (3.2%), Hispanic; 278 (0.7%), North American Native; 29 986 (78.1%), White; 373 (1.0%), of other race or ethnicity; and 404 (1.1%), of unknown race or ethnicity. Compared with patients undergoing gastric bypass, at 5 years after surgery, patients undergoing sleeve gastrectomy had a lower cumulative incidence of mortality (4.27%; 95% CI, 4.25%-4.30% vs 5.67%; 95% CI, 5.63%-5.69%), complications (22.10%; 95% CI, 22.06%-22.13% vs 29.03%; 95% CI, 28.99%-29.08%), and reintervention (25.23%; 95% CI, 25.19%-25.27% vs 33.57%; 95% CI, 33.52%-33.63%). Conversely, patients undergoing sleeve gastrectomy had a higher cumulative incidence of surgical revision at 5 years (2.91%; 95% CI, 2.90%-2.93% vs 1.46%; 95% CI, 1.45%-1.47%). The adjusted hazard ratio (aHR) of all-cause hospitalization and ED use was lower for patients undergoing sleeve gastrectomy at 1 year (hospitalization, aHR, 0.83; 95% CI, 0.80-0.86; ED use, aHR, 0.87; 95% CI, 0.84-0.90) and 3 years (hospitalization, aHR, 0.94; 95% CI, 0.90-0.98; ED use, aHR, 0.93; 95% CI, 0.90-0.97) after surgery but similar between groups at 5 years (hospitalization, aHR, 0.99; 95% CI, 0.94-1.04; ED use, aHR, 0.97; 95% CI, 0.92-1.01). Total health care spending among patients undergoing sleeve gastrectomy was lower at 1 year after surgery ($28 706; 95% CI, $27 866-$29 545 vs $30 663; 95% CI, $29 739-$31 587), but similar between groups at 3 ($57 411; 95% CI, $55 239-$59 584 vs $58 581; 95% CI, $56 551-$60 611) and 5 years ($86 584; 95% CI, $80 183-$92 984 vs $85 762; 95% CI, $82 600-$88 924). Conclusions and relevance: In a large cohort of patients undergoing bariatric surgery, sleeve gastrectomy was associated with a lower long-term risk of mortality, complications, and reinterventions but a higher long-term risk of surgical revision. Understanding the comparative safety of these operations may better inform patients and surgeons in their decision-making.
Article
Background Laparoscopic surgery is regarded as the gold standard for the surgical management of cholelithiasis. To improve post-operative pain, low-pressure laparoscopic cholecystectomies (LPLC) have been trialed. A recent systematic review found that LPLC reduced pain; however, many of the randomised control trials were at a high risk of bias and the overall quality of evidence was low.Methods One hundred patients undergoing elective laparoscopic cholecystectomy were randomised to a LPLC (8 mmHg) or a standard pressure laparoscopic cholecystectomy (12 mmHg) (SPLC) with surgeons and anaesthetists blinded to the pressure. Pressures were increased if vision was compromised. Primary outcomes were post-operative pain and analgesia requirements at 4–6 h and 24 h.ResultsIntra-operative visibility was significantly reduced in LPLC (p<0.01) resulting in a higher number of operations requiring the pressure to be increased (29% vs 8%, p=0.010); however, there were no differences in length of operation or post-operative outcomes. Pain scores were comparable at all time points across all pressures; however, recovery room fentanyl requirement was more than four times higher when comparing 8 to 12 mmHg (12.5mcg vs 60mcg, p=0.047). Nausea and vomiting was also higher when comparing these pressures (0/36 vs 7/60, p=0.033). Interestingly, when surgeons estimated the operating pressure, they were correct in only 69% of cases.Conclusion Although pain scores were similar, there was a significant reduction in fentanyl requirement and nausea/vomiting in LPLC. Although LPLC compromised intra-operative visibility requiring increased pressure in some cases, there was no difference in complications, suggesting LPLC is safe and beneficial to attempt in all patients.Trial RegistrationRegistered with the Australia and New Zealand Clinical Trials Registry (ACTRN12619000205134).