You are on page 1of 7

The Effect of Intraperitoneal Local Anesthesia in

Laparoscopic Cholecystectomy: A Systematic Review


and Meta-Analysis
Alexander P. Boddy, BM, BCh Intraperitoneal administration of local anesthesia is often used to improve pain
relief after laparoscopic cholecystectomy. We have conducted a meta-analysis to
Samir Mehta, BM, BCh establish the efficacy of this technique in reducing early postoperative abdominal
pain. A systematic literature search revealed 24 randomized, controlled trials
assessing intraperitoneal local anesthetic use in laparoscopic cholecystectomy that
Michael Rhodes, MD
Downloaded from http://journals.lww.com/anesthesia-analgesia by BhDMf5ePHKav1zEoum1tQfN4a+kJLhEZgbsIHo4XMi0hCywCX1AWnYQp/IlQrHD3i3D0OdRyi7TvSFl4Cf3VC4/OAVpDDa8K2+Ya6H515kE= on 03/31/2021

met inclusion criteria. Of these, 16 studies reported sufficient data to allow pooled
quantitative analysis. The weighted mean differences (WMD) in visual analog pain
score at 4 h after surgery were pooled using a random effects model. Overall, the
use of intraperitoneal local anesthesia resulted in a significantly reduced pain score
at 4 h (WMD, ⫺9 mm; 95% confidence interval [CI], ⫺13 to ⫺5). Subgroup analysis
suggested that the effect was greater when the local anesthetic was given at the
start of the operation (WMD, ⫺13 mm; 95% CI, ⫺19 to ⫺7) compared with
instillation at the end (WMD, ⫺6 mm; 95% CI, ⫺10 to ⫺2). No adverse events
related to local anesthetic toxicity were reported. We conclude that the use of
intraperitoneal local anesthesia is safe, and it results in a statistically significant
reduction in early postoperative abdominal pain.
(Anesth Analg 2006;103:682–8)

L aparoscopic cholecystectomy is the treatment of


choice for symptomatic cholelithiasis. Although there
have reported no benefit. We have performed a sys-
tematic review of the literature to evaluate the effects
are clear benefits compared with open surgery, post- of intraperitoneal local analgesia on early postopera-
operative pain after laparoscopic cholecystectomy re- tive abdominal pain after laparoscopic cholecystec-
mains an issue. Pain can prolong hospital stay and tomy.
lead to increased morbidity, which is particularly
important now that many centers are performing this METHODS
operation as a day-case procedure. We systematically identified reports of random-
Administration of intraperitoneal local anesthetic ized, controlled trials assessing the use of intraperito-
(LA), either during or after surgery, is used by many neal LA in the setting of laparoscopic cholecystec-
surgeons as a method of reducing postoperative pain. tomy. MEDLINE, EMBASE, and the Cochrane Library
This technique was first evaluated in patients under- databases were searched in June 2005 using the term:
going gynecological laparoscopic surgery (1). Its ap- laparoscopic cholecystectomy AND intraperitoneal
plication in laparoscopic cholecystectomy was initially AND [local an(a)esthetic OR local an(a)esthesia OR
examined in a randomized trial in 1993 (2). Since then, lidocaine OR lignocaine OR bupivacaine OR levobupi-
many trials evaluating the efficacy of intraperitoneal vacaine OR ropivacaine]. No language restrictions
were imposed. Additional reports were identified
LA in laparoscopic cholecystectomy have been pub-
from reference lists of retrieved papers.
lished worldwide. Although a number of these studies
Studies included were all double-blind, random-
have reported a significant reduction in postoperative
ized comparisons of intraperitoneal LA versus placebo
pain after the use of intraperitoneal analgesia, others
or no treatment, evaluating abdominal pain in the
setting of laparoscopic cholecystectomy. Trials com-
bining intraperitoneal LA with other interventions
From the Department of General Surgery, Norfolk and Norwich (e.g., port site infiltration or intraperitoneal nonsteroi-
University Hospital, United Kingdom.
dal antiinflammatory drugs) were included as long as
Accepted for publication April 25, 2006.
there were comparable treatment and control groups
Address correspondence and reprint requests to M. Rhodes,
MD, Consultant Surgeon, Norfolk and Norwich University Hospi- in which the only difference was instillation of intra-
tal, Colney Lane, Norwich, Norfolk, NR4 7UY, United Kingdom. peritoneal LA. Trials in which infusions of LA were
Address e-mail to mr@lapsurgeon.co.uk. administered after the patient had recovered from
Copyright © 2006 International Anesthesia Research Society anesthesia were excluded. Papers that could not be
DOI: 10.1213/01.ane.0000226268.06279.5a
supplied by the British Library were not included. The

682 Vol. 103, No. 3, September 2006


literature search, determination of studies meeting the Because there was considerable clinical heterogene-
inclusion criteria, and data extraction were performed ity among trials (different quantities and concentra-
independently by two authors. Consensus was tions of different LAs were used and different postop-
reached by discussion. erative analgesia regimens were used), pooled
The methodological quality of each eligible study analysis was performed using a random effects model.
was assessed using a 3-item, 5-point scale, which has Studies that involved routine patient-controlled anal-
previously been validated (3). Studies described as gesia (PCA) regimens were assessed in a separate
randomized were given either 1 or 2 points if the subgroup because it was expected that these patients
method of randomization was described and was would have less postoperative pain than patients who
appropriate. One point was deducted if randomiza- needed to ask the nursing staff for each dose of
tion was inappropriate. Studies described as double- analgesia.
blind were either given 1 or 2 points if the method of
blinding was described and was appropriate. One
RESULTS
point was deducted if blinding was inappropriate. If
The initial electronic literature searches revealed 59
the numbers were described and reasons for with-
studies, and after review of the abstracts, 31 random-
drawals offered, a further point was given. As only
ized trials were identified as potentially meeting the
randomized, double-blind trials were included, the
inclusion criteria. Of these, we were unable to retrieve
minimum possible score for each study was 2 and the
the full papers of two trials (5,6). Four studies were
maximum 5.
excluded because there were differences between
The principal outcome measure for quantitative
treatment and control arms aside from the instillation
analysis was abdominal pain score at 4 h after surgery.
of intraperitoneal local analgesia (7–10), and one study
Only studies reporting pain scores on a visual analog
was excluded because it examined the use of a post-
scale (VAS) (0 –100 mm or 0 –10 cm) or an equivalent
operative infusion of intraperitoneal local analgesia
verbal pain score (0 –10) were included. If both were (11).
given, the VAS was used in preference. Whereas many Twenty-four studies were incorporated in this sys-
studies provided just one overall pain score, several tematic review (2,12–34) (Table 1). Anesthetic drugs
studies reported pain scores at rest, with movement or that were evaluated included bupivacaine, levobupi-
coughing, and for visceral abdominal pain, superficial vacaine, lidocaine, and ropivacaine. In some studies,
pain, and shoulder pain. In these cases, the pain score bupivacaine or levobupivacaine was used in conjunc-
that best represented abdominal pain at rest was used. tion with adrenaline (epinephrine). Most studies used
Most studies reported pain scores at various times a set dose and concentration of LA. However, some
during the postoperative period. If the score at 4 h was studies based the quantity of LA used on the patient’s
not reported, the nearest time point to 4 h was used weight. In these cases, the figures given in Table 1 are
(between 1 and 6 h). Mean pain scores and standard based on the average weight of patients in the treat-
deviations for the control and intervention groups ment arm. In one study, the total quantity of LA used
were extracted, either from the text or from tables or was stated, but no mention was made of the strength
graphs included within the report. If the mean score or volume of the solution (23). The timing and location
was not given, the median score was taken as an of instillation of local analgesia are also shown in
approximation to the mean. In some cases, an estima- Table 1.
tion of standard deviation was made using Cochrane
review methodology (4). Studies offering no measure Pain Scores
of dispersion were not included in the pooled quanti- Twelve of the 24 studies reported a significant
tative analysis. The weighted mean difference (WMD) improvement in pain during the early postoperative
between the treatment and control groups was calcu- period. In one study, this occurred for pain during
lated using Review Manager 4.2.8 software (The Co- inspiration but not for pain at rest (34). In another
chrane Collaboration, Oxford, United Kingdom). The study, no significant improvement in pain was noticed
WMD is reported in millimeters (the units of the VAS in the treatment arms that received a single bolus
for pain), and a negative value represents lower pain injection of LA, but a 4-h postoperative infusion did
scores in patients in the treatment groups compared to result in significantly lower VAS pain scores (30).
the control groups. Four studies (18,26,30,32) had treatment arms in
Quantitative analysis was also performed on data which the LA was instilled at different times in
given for additional analgesia requirements. The mean relation to surgery. However, only one of these studies
and standard deviation of the additional analgesia reported a significant difference in pain scores be-
requirements (in milligrams) for the control and inter- tween groups at different times (18). In this study,
vention groups were extracted from each report. Be- pain scores were lower in the group receiving LA
cause different analgesic drugs with various potencies before surgery than the group receiving LA after
were used, comparison among studies required stan- surgery. A third group receiving LA both before and
dardized mean differences to be calculated. after surgery had even lower scores.

Vol. 103, No. 3, September 2006 © 2006 International Anesthesia Research Society 683
Table 1. Included Studies
Included in
LA used quantitative analysis?

Timing of Location
Patients Total LA in of LA
Quality (control/ Volume Strength quantity Relation to instil- PCA VAS at Postop Significant
Study and year score treatment) Type (mL) (%) (mg) dissection lation used? 1– 6 h analgesia difference

Chundrigar, 1993 (2) 3 30/28 B 20 0.25 50 after GBB No No No Yes


Rademaker, 1994 (12) 2 15/15 B 20 0.25 50 after SD No Yes Yes No
Joris, 1995 (13) 2 20/20 BA 80 0.125 100 after SD No Yes No No
Raetzell, 1995 (14) 2 12/12 B 50 0.25 125 after SD, GBB Yes Yes Yes No
Scheinin, 1995 (15) 2 20/20 B 100 0.15 150 after SD No No Yes No
Fornari, 1996 (16) 2 50/50 BA 60 0.16 96 after SD, GIP No No No No
Fuhrer, 1996 (17) 3 12/12 B 48 0.375 180 after SD, GBB Yes Yes Yes No
Pasqualucci, 1996 (18) 3 27/28 BA 40 0.5 200 before, after, SD, GBB No Yes Yes Yes
before and aftera
Szem, 1996 (19) 3 29/26 B 100 0.1 100 before SD, GBB No Yes Yes Yes
Mraovic, 1997 (20) 4 40/40 B 30 0.5 150 before and after SD, GBB No Yes Yes Yes
Weber, 1997 (21) 2 50/50 B 10 0.5 120 after SD No No No Yes
Tsimoyiannis, 1998 (22) 3 50/50 B 48 0.25 120 after SD No Yes Yes Yes
Elfberg, 2000 (23) 3 32/33 B ns ns 154 after GBB No Yes No No
Kolsi, 2000 (24) 4 20/20 L 40 1 400 before SD, GBB No Yes No Yes
Zmora, 2000 (25) 4 25/26 B 50 0.2 100 after SD, GBB No No No No
Lee, 2001 (26) 3 25/20 B 40 0.25 100 beforea, after SD, GBB No No No No
Jiranantarat, 2002 (27) 2 41/39 B 20 0.5 100 after SD, GBB No Yes Yes No
Labaille, 2002 (28) 3 12/11 R 40 0.75 300 before and after SD, GBB Yes Yes Yes Yes
Maestroni, 2002 (29) 3 30/30 R 200 0.175 350 before (pneu- GIP No Yes No Yes
moperitoneum)
Karadeniz, 2003 (30) 3 15/15 B 20 0.5 100 before, aftera SD, GBB No No No No (only for
infusion)
Lepner, 2003 (31) 5 20/20 L 200 0.15 300 after SD No Yes No No
Paulson, 2003 (32) 3 14/19 B 30 0.5 150 before, after, SD, GBB No No No Yes
before and aftera
Razek, 2003 (33) 4 20/20 LB 60 0.25 150 after SD, GBB No Yes No Yes
Ng, 2004 (34) 5 22/21 LBA 30 0.25 75 after SD, GBB No Yes No Yes (during
inspiration only)
LA ⫽ local anesthetic; PCA ⫽ patient-controlled analgesia; ns ⫽ not stated; L ⫽ lignocaine (lidocaine); B ⫽ bupivacaine; LB ⫽ levobupivacaine; R ⫽ ropivacaine; A ⫽ adrenaline (epinephrine);
SD ⫽ sub-diaphragmatic; GBB ⫽ gallbladder bed (or over gallbladder if before dissection); GIP ⫽ generalized intraperitoneal.
a
Time had most significant effect.

Many of the studies reported additional analgesic 34). In the postoperative period, three studies used
use by patients, either as the number of patients PCA IV regimens (14,17,28), whereas in the other
requiring additional analgesia (15,16,20,22,29,31,34), studies, patients were provided with analgesia by the
the time to first analgesia request (27), the number of nursing staff.
requests for analgesia (13), the number of doses of Eight studies measured plasma levels after intra-
analgesic (25), or the mean and total dose of a single or peritoneal administration of LA in 161 patients. Three
combination of analgesic drugs (2,12,14,15,17–22,27,28, of these trials studied bupivacaine (14,15,17), one

Figure 1. Effect of intraperitoneal instillation of local anesthetics (LAs) on early postoperative pain.
684 Intraperitoneal Local Anesthesia in Lap Chole ANESTHESIA & ANALGESIA
Figure 2. Effect of timing of intraperitoneal instillation of local anesthetics (LA) on early postoperative pain.

levobupivacaine (33), one lidocaine (24), two ropiva- 4-point scale for recording pain and was therefore not
caine (28,29), and one both lidocaine and bupivacaine included in any quantitative analysis (15).
(12). Potentially toxic plasma levels were reported in 4 Sixteen of the included studies reported results
patients overall: 1 patient after 50 mL of 0.25% bupiv- such that sufficient data could be extracted for quan-
acaine (125 mg) (14), 1 patient after 0.6 mL/kg of titative analysis (12–14,17–20,22–24,27–29,31,33,34).
0.375% bupivacaine (17), and 2 patients after 40 mL of Only one treatment arm per trial was included in the
0.75% ropivacaine (300 mg) (28). However, no patient initial meta-analysis: if the trial compared different
in any of the trials included in this review suffered any Las, different doses of the same LA, or different times
adverse event attributable to the use of LA. at which the LA was instilled, the arm in which the
Two studies examined the effect of intraperitoneal effect was greatest was used for the pooled analysis. In
LA on length of hospital stay. One reported that the total, there were 397 patients in the treatment arms
use of intraperitoneal bupivacaine did not affect the and 400 patients in the control arms. There was a
length of hospitalization (15), but another, by combin- statistically significant overall WMD in VAS scores of
ing treatment arms, reported a significant increase in –9 mm (95% confidence intervals [CI], ⫺13 to ⫺5) in
the proportion of patients able to be discharged on the favor of treatment (Fig. 1). As expected, there was a
same day as surgery (32). significant degree of heterogeneity among the studies,
Two studies assessed respiratory function as an as demonstrated by an I2 value of 74.8% (I2 is a
outcome measure. One found no difference in peak measure used to quantify heterogeneity and repre-
expiratory flow rates between patients receiving intra- sents the percentage of the variability that is caused by
peritoneal bupivacaine and controls (23). However, heterogeneity rather than sampling error: a value
the second study found that intraperitoneal lidocaine more than 50% may be considered to represent sub-
significantly reduced forced vital capacity at 4 h after stantial heterogeneity).
surgery and increased hypoxemic periods in the 6 h To further examine the effects of timing of instilla-
after surgery when compared with controls (14). tion, the pooled quantitative analysis was repeated,
All of the included studies used postoperative pain grouping the studies according to when LA was used.
as an outcome measure. However, one study used a Studies in which LA was instilled at the end of surgery

Vol. 103, No. 3, September 2006 © 2006 International Anesthesia Research Society 685
Figure 3. Effect of intraperitoneal instillation of local anesthetics (LA) on postoperative analgesia requirements.

were placed into Subcategory 1. Studies in which LA a meta-analysis (35) published in 2000. This previous
was instilled before any dissection (19,29), or in which review reported improved pain relief in 7 of 13 trials
there were two instillations of local analgesia— one at and a meta-analysis of 10 trials found an overall WMD
the beginning and one at the end of surgery in VAS of ⫺13 mm in favor of the treatment groups.
(20,28)—were placed into Subcategory 2. In one study, However, we did not find a significant effect of
data were available that were applicable to both intraperitoneal LA on the total amount of analgesia
subcategories (18). The WMD in VAS scores for Sub- delivered in the postoperative period. This might be
category 1 (LA after surgery) was ⫺6 mm (95% CI, explained by the fact that LA has its effects only over
⫺10 to ⫺2), whereas the WMD for Subcategory 2 (LA the initial few hours. In many of the studies, delivery
before surgery) was ⫺13 mm (95% CI, ⫺19 to ⫺8) (Fig. of analgesia was measured over periods far in excess
2). This gives a significant difference in WMD between of this timescale.
these subcategories of 7.0 mm in favor of LA before The results of this meta-analysis highlight the con-
surgery (approximate 95% CI, 0.1–14.0). siderable heterogeneity of results from available trials.
The 10 studies that reported doses of additional Some of the factors that may be responsible for the
postoperative analgesia to enable quantitative analysis clinical heterogeneity among trials are summarized in
are identified in Table 1. Pooled analysis of these Table 2. These factors may either directly influence the
studies was performed as shown in Figure 3. Overall, efficacy of the LA or may affect postoperative pain
the standardized mean differences in analgesia use independently (36), thus reducing the potential benefit
between treatment and control arms was not signifi- from the administration of intraperitoneal LA. For
cant at ⫺0.77 (95% CI, ⫺1.65– 0.12). example, patients in trials where PCA was used
generally had lower pain scores than patients who had
DISCUSSION to request each dose of analgesia from medical or
Laparoscopic cholecystectomy is one of the most nursing staff. This may explain why, in trials where
frequently performed elective general surgical opera- PCA was used, intraperitoneal LA resulted in a
tions. It is an ideal candidate to be performed as a smaller reduction in early postoperative pain than in
day-case or short-stay procedure, and therefore, the
provision of adequate postoperative pain relief is of
Table 2. Factors That May Influence the Benefits of
considerable importance. Instillation of intraperito- Intraperitoneal Anesthesia
neal LA to reduce postoperative pain has been studied
through randomized trials for more than 10 years, and Factor
this review has collated the available data both quali- Dose and concentration of local anesthetic used
tatively and quantitatively. Site of instillation (sub-diaphragmatic versus sub-hepatic)
We identified 24 studies that were suitable for Timing of instillation (before versus after surgery)
qualitative analysis. In half of these, there was a Pneumoperitoneum (volume, pressure, and temperature
of)
significant improvement in postoperative pain relief Volume of residual CO2 (causing diaphragmatic irritation)
after instillation of intraperitoneal LA. Meta-analysis Spillage of bile and blood (may interfere with absorption)
revealed an overall WMD in VAS of ⫺9 mm in favor Degree of nonvisceral pain (e.g., from incision sites)
of the treatment groups. Although statistically signifi- Instillation in head-down position versus supine
cant, this is slightly lower than the difference found in Postoperative analgesia regimen

686 Intraperitoneal Local Anesthesia in Lap Chole ANESTHESIA & ANALGESIA


trials not using PCA (WMD, ⫺6 versus ⫺10 mm).
However, we did not find that patients who received
intraperitoneal LA used a significantly smaller total
dose of PCA than control patients.
As well as differences in surgical and anesthetic
technique, there were also differences in pain out-
comes that different studies tried to measure. Al-
though many studies reported only an overall ab-
dominal pain score, several asked patients to
distinguish visceral pain from superficial abdominal
pain (2,13,26,28) or shoulder-tip pain (13,16,21,22,
26,28,31,34) and also measured pain on movement
(8,13,15,28), coughing (8,13,14,28), deep inspiration
(14,34), and at rest. Although we tried to extract
comparable data for the pooled quantitative analysis,
these differences may have had an influence on the
heterogeneity of the meta-analysis. Figure 4. Relationship between strength (concentration) of
Some authors have suggested that the timing of LA local anesthetic (LA) and early postoperative mean differ-
administration has an important role in the success of ence in Visual Analog Scale (VAS) pain score. Unweighted,
the technique (18,29,37). It has been argued that post- linear regression of all trials evaluating bupivacaine or
operative pain is reduced if suppression of central levobupivacaine (identified by reference number) was per-
formed using SPSS 13.0 for Windows (SPSS Inc, Chicago,
neural sensitization by intraperitoneal LA occurs be- IL).
fore nociceptive stimuli have triggered the activation
of pain pathways, compared with afterwards. Pooled
analyses seemed to support this view: the WMD in
in this study, significantly more patients who received
VAS scores for studies in which LA was only admin-
intraperitoneal LA were discharged on the day of the
istered at the end of surgery was smaller than for
procedure (79% versus 43%; P ⬍ 0.02) (32).
studies in which at least some LA was administered
Overall, this review does lend limited support to
before any dissection took place.
the use of intraperitoneal LA in laparoscopic cholecys-
There is little evidence with regard to which type of
tectomy as part of a multimodal approach to pain
LA is most effective because limited data are available
management. The technique seems to be safe and
for drugs other than bupivacaine. Bupivacaine itself
results in a statistically significant reduction in early
(or levobupivacaine) is an excellent choice for intra-
postoperative abdominal pain. It may be of particular
peritoneal LA because of its long duration of action.
benefit when the operation is planned as an ambula-
Linear regression analysis of the VAS pain scores from
tory procedure to improve same-day discharge rates.
all trials using bupivacaine or levobupivacaine sug-
Finally, there is some evidence to suggest that LA may
gested that there was a significant correlation (P ⫽
be more effective if used at a larger strength and if at
0.02; R2 ⫽ 0.32) between the strength of bupivacaine
least some is instilled before any dissection.
used and difference in pain score between treatment
and control groups, i.e., larger concentrations of bu-
REFERENCES
pivacaine resulted in larger reductions in pain score
(Fig. 4). However, there was no significant correlation 1. Narchi P, Benhamou D, Fernandez H. Intraperitoneal local
anaesthetic for shoulder pain after day-case laparoscopy. Lancet
between the volume or total quantity of LA used and 1991;338:1569–70.
the WMD in pain score. 2. Chundrigar T, Hedges AR, Morris R, Stamatakis JD. Intraperi-
In this study, we calculated a WMD of 9 mm in toneal bupivacaine for effective pain relief after laparoscopic
cholecystectomy. Ann R Coll Surg Engl 1993;75:437–9.
favor of intraperitoneal instillation of LA. Although 3. Jadad AR, Moore RA, Carroll D, et al. Assessing the quality of
statistically significant, the clinical significance of this reports of randomized clinical trials: is blinding necessary?
difference in pain perception is uncertain. Although 9 Control Clin Trials 1996;17:1–12.
4. Alderson P, Green S, Higgins JPT, et al. Cochrane reviewers’
mm is less than the difference of 13 mm on a 0- to handbook 4.2.3 [updated November 2004]. In: The Cochrane
100-mm VAS for acute pain that has been suggested to Library. Chichester, UK: John Wiley & Sons, Ltd.
be clinically significant (38,39), it could be argued that 5. Kilic A, Basgul E, Ozdemir A, Erdem MK. [The efficacy of the
intraperitoneal bupivacaine application on early postoperative
any reduction in pain is beneficial for the patient if pain and blood gas values after laparoscopic cholecystectomy].
there are no adverse effects associated with it. In Agri Derg 1996;8:20–6.
particular, a smaller difference in pain may be more 6. Rodrigues-Moreira W, Amatto PE, Cardoso-Bessa RJ, Belchior-
Silva J. [Intraperitoneal bupivacaine for pain relief after laparo-
significant for patients whose baseline VAS is rela- scopic cholecystectomy]. Rev Bras Anestesiol 2000;50:283–8.
tively low (40) and may determine the difference 7. Bisgaard T, Klarskov B, Kristiansen VB, et al. Multi-regional
between day-case discharge and an overnight stay in local anesthetic infiltration during laparoscopic cholecystec-
tomy in patients receiving prophylactic multi-modal analgesia:
hospital. We identified only one trial that reported a a randomized, double-blinded, placebo-controlled study.
same-day discharge rate as an outcome measure, and Anesth Analg 1999;89:1017–24.

Vol. 103, No. 3, September 2006 © 2006 International Anesthesia Research Society 687
8. Elhakim M, Amine H, Kamel S, Saad F. Effects of intraperitoneal randomized, placebo-controlled and double-blind study. Surg
lidocaine combined with intravenous or intraperitoneal tenoxi- Laparosc Endosc Percutan Tech 2000;10:357–9.
cam on pain relief and bowel recovery after laparoscopic 24. Kolsi K, Ghozzi H, Masmoudi A, et al. Intraperitoneal ligno-
cholecystectomy. Acta Anaesthesiol Scand 2000;44:929–33. caine for analgesia after laparoscopic cholecystectomy. Acute
9. Hernandez-Palazon J, Tortosa JA, Nuno de la Rosa V. Intraperi- Pain 2000;3:200–5.
toneal application of bupivacaine plus morphine for pain relief 25. Zmora O, Stolik-Dollberg O, Bar-Zakai B, et al. Intraperitoneal
after laparoscopic cholecystectomy. Eur J Anaesthesiol bupivacaine does not attenuate pain following laparoscopic
2003;20:891–6. cholecystectomy. Jsls 2000;4:301–4.
10. Schulte-Steinberg H, Weninger E, Jokisch D, et al. Intraperito- 26. Lee IO, Kim SH, Kong MH, et al. Pain after laparoscopic
neal versus interpleural morphine or bupivacaine for pain after cholecystectomy: the effect and timing of incisional and intra-
laparoscopic cholecystectomy. Anesthesiology 1995;82:634–40. peritoneal bupivacaine. Can J Anaesth 2001;48:545–50.
11. Gupta A, Thorn SE, Axelsson K, et al. Postoperative pain relief 27. Jiranantarat V, Rushatamukayanunt W, Lert-akyamanee N.
using intermittent injections of 0.5% ropivacaine through a Analgesic effect of intraperitoneal instillation of bupivacaine for
catheter after laparoscopic cholecystectomy. Anesth Analg postoperative laparoscopic cholecystectomy. J Med Assoc Thai
2002;95:450–6. 2002;85:S897–903.
12. Rademaker BM, Kalkman CJ, Odoom JA, et al. Intraperitoneal 28. Labaille T, Mazoit JX, Paqueron X, et al. The clinical efficacy and
local anaesthetics after laparoscopic cholecystectomy: effects on pharmacokinetics of intraperitoneal ropivacaine for laparo-
postoperative pain, metabolic responses and lung function. Br J scopic cholecystectomy. Anesth Analg 2002;94:100–5.
Anaesth 1994;72:263–6. 29. Maestroni U, Sortini D, Devito C, et al. A new method of
13. Joris J, Thiry E, Paris P, et al. Pain after laparoscopic cholecys- preemptive analgesia in laparoscopic cholecystectomy. Surg
tectomy: characteristics and effect of intraperitoneal bupiva- Endosc 2002;16:1336–40.
caine. Anesth Analg 1995;81:379–84. 30. Karadeniz Ü, Erdemli Ö, Ünver S, et al. Intraperitoneal bupiv-
14. Raetzell M, Maier C, Schroder D, Wulf H. Intraperitoneal acaine infusion and injection for postoperative pain manage-
application of bupivacaine during laparoscopic cholecystec- ment in laparoscopic cholecystectomy. Anestezi Dergisi
2003;11:226–30.
tomy: risk or benefit? Anesth Analg 1995;81:967–72.
31. Lepner U, Goroshina J, Samarutel J. Postoperative pain relief
15. Scheinin B, Kellokumpu I, Lindgren L, et al. Effect of intraperi-
after laparoscopic cholecystectomy: a randomised prospective
toneal bupivacaine on pain after laparoscopic cholecystectomy.
double-blind clinical trial. Scand J Surg 2003;92:121–4.
Acta Anaesthesiol Scand 1995;39:195–8.
32. Paulson J, Mellinger J, Baguley W. The use of intraperitoneal
16. Fornari M, Miglietta C, Di Gioia S, et al. [The use of intraopera-
bupivacaine to decrease the length of stay in elective laparo-
tive topical bupivacaine in the control of postoperative pain
scopic cholecystectomy patients. Am Surg 2003;69:275–8.
following laparoscopic cholecystectomy]. Minerva Chir 33. Razek EA. Intraperitoneal levobupivacaine for analgesia after
1996;51:881–5. laparoscopic cholecystectomy. Egyptian J Anaesth 2003;19:
17. Fuhrer Y, Charpentier C, Boulanger G, et al. [Analgesia after 249–54.
laparoscopic cholecystectomy by intraperitoneal administration 34. Ng A, Swami A, Smith G, et al. Is intraperitoneal levobupiva-
of bupivacaine]. Ann Fr Anesth Reanim 1996;15:128–34. caine with epinephrine useful for analgesia following laparo-
18. Pasqualucci A, de Angelis V, Contardo R, et al. Preemptive scopic cholecystectomy: a randomized controlled trial. Eur J
analgesia: intraperitoneal local anesthetic in laparoscopic Anaesthesiol 2004;21:653–7.
cholecystectomy—a randomized, double-blind, placebo- 35. Moiniche S, Jorgensen H, Wetterslev J, Dahl JB. Local anesthetic
controlled study. Anesthesiology 1996;85:11–20. infiltration for postoperative pain relief after laparoscopy: a
19. Szem JW, Hydo L, Barie PS. A double-blinded evaluation of qualitative and quantitative systematic review of intraperito-
intraperitoneal bupivacaine vs saline for the reduction of post- neal, port-site infiltration and mesosalpinx block. Anesth Analg
operative pain and nausea after laparoscopic cholecystectomy. 2000;90:899–912.
Surg Endosc 1996;10:44–8. 36. Wills VL, Hunt DR. Pain after laparoscopic cholecystectomy.
20. Mraovic B, Jurisic T, Kogler-Majeric V, Sustic A. Intraperitoneal Br J Surg 2000;87:273–84.
bupivacaine for analgesia after laparoscopic cholecystectomy. 37. Lagergren J, Bergstrom R, Lindgren A, Nyren O. Symptomatic
Acta Anaesthesiol Scand 1997;41:193–6. gastroesophageal reflux as a risk factor for esophageal adeno-
21. Weber A, Munoz J, Garteiz D, Cueto J. Use of subdiaphragmatic carcinoma. N Engl J Med 1999;340:825–31.
bupivacaine instillation to control postoperative pain after lapa- 38. Todd KH, Funk KG, Funk JP, Bonacci R. Clinical significance of
roscopic surgery. Surg Laparosc Endosc 1997;7:6–8. reported changes in pain severity. Ann Emerg Med 1996;27:
22. Tsimoyiannis EC, Glantzounis G, Lekkas ET, et al. Intraperito- 485–9.
neal normal saline and bupivacaine infusion for reduction of 39. Gallagher EJ, Liebman M, Bijur PE. Prospective validation of
postoperative pain after laparoscopic cholecystectomy. Surg clinically important changes in pain severity measured on a
Laparosc Endosc 1998;8:416–20. visual analog scale. Ann Emerg Med 2001;38:633–8.
23. Elfberg BA, Sjovall-Mjoberg S. Intraperitoneal bupivacaine does 40. Bird SB, Dickson EW. Clinically significant changes in pain
not effectively reduce pain after laparoscopic cholecystectomy: a along the visual analog scale. Ann Emerg Med 2001;38:639–43.

688 Intraperitoneal Local Anesthesia in Lap Chole ANESTHESIA & ANALGESIA

You might also like