SlideShare a Scribd company logo
1 of 6
Download to read offline
American Journal of Emergency Medicine (2007) 25, 385 – 390




                                                                                                                    www.elsevier.com/locate/ajem



Original Contribution

Management of severe acute pain in emergency settings:
ketamine reduces morphine consumptionB
Michel Galinski MDa,*, Francois Dolveck MDb, Xavier Combes MDe,
                           ¸
Veronique Limoges MD , Nadia Smail MD, PhDd, Veronique Pommier MDc,
 ´                     c           !!
                                                 ´
                     b                   a
Francois Templier MD , Jean Catineau MD ,
    ¸
Frederic Lapostolle MDa, Frederic Adnet MD, PhDa
  ´ ´                      ´ ´
a
 Samu 93-EA 3409 Avicenne Hospital, 93009 Bobigny Cedex, France
b
  Samu 92-Raymond Poincare Hospital, 92100 Garches, France
                           ´
c
 Emergency Department-Smur 77, Meaux’s Hospital, 77100 Meaux, France
d
  Samu 31-Purpan Hospital, 31000 Toulouses, France
e
                                          ´
 Samu 94-Henri Mondor Hospital, 94000 Creteil, France

Received 1 July 2006; revised 1 November 2006; accepted 2 November 2006


Abstract
Objective: The aim of the study was to compare in emergency settings 2 analgesic regimens, morphine
with ketamine (K group) or morphine with placebo (P group), for severe acute pain in trauma patients.
Methods: This was a prospective, multicenter, randomized, double-blind, clinical trial. Seventy-three
trauma patients with a severe acute pain defined as a visual analog scale (VAS) score of at least 60/100
were enrolled. Patients in the K group received 0.2 mg d kgÀ1 of intravenous ketamine over 10 minutes,
and patients in the P group received isotonic sodium chloride solution. In both groups, patients were
given an initial intravenous morphine injection of 0.1 mg d kgÀ1, followed by 3 mg every 5 minutes.
Efficient analgesia was defined as a VAS score not exceeding 30/100. The primary end points were
morphine consumption and VAS at 30 minutes (T30).
Results: At T30, morphine consumption was significantly lower in the K group vs the P group, with
0.149 mg d kgÀ1 (0.132-0.165) and 0.202 mg d kgÀ1 (0.181-0.223), respectively ( P b .001). The VAS
score at T30 did not differ significantly between the 2 groups, with 34.1 (25.6-42.6) in the K group and
39.5 (32.4-46.6) in the P group ( P = not significant).
Conclusion: Ketamine was able to provide a morphine-sparing effect.
D 2007 Elsevier Inc. All rights reserved.




                                                                              1. Introduction
   Presented at the European Society of Anaesthesiology Congress in
Madrid, Spain, June 3-6, 2006.
   B                                                                             Morphine titration infusion usually provides rapid and
      Support was provided solely by institutional and/or departmental
sources.                                                                      effective analgesia in severe acute pain [1]. However,
   * Corresponding author. Tel.: +33 1 48 96 44 55; fax: +33 1 48 96 44 45.   adverse effects sometimes occur and may require discon-
   E-mail address: michel.galinski@avc.ap-hop-paris.fr (M. Galinski).         tinuation of morphine titration before sufficient pain relief is

0735-6757/$ – see front matter D 2007 Elsevier Inc. All rights reserved.
doi:10.1016/j.ajem.2006.11.016
386                                                                                                           M. Galinski et al.

obtained [2]. The combination of nonopioid analgesics with        randomly allocated to receive either morphine and ket-
morphine provides a morphine-sparing effect and should            amine (K group) or morphine and placebo (P group).
decrease toxicity. This concept is the basis of multimodal        Patients were eligible for inclusion if they presented a
analgesia [3].                                                    trauma with a severe acute pain defined as a visual analog
    Small doses of ketamine possess N-methyl-d-aspartate          scale (VAS) score of at least 60/100; were aged between
(NMDA) receptor noncompetitive antagonist properties              18 and 70 years; and were without acute respiratory,
through a magnesium-dependent channel blockade [4].               hemodynamic, or neurologic compromise (respiratory
Several studies demonstrated that it improves opioid              distress signs, systolic blood pressure V90 mm Hg,
analgesia in postoperative settings [5,6].                        Glasgow Coma Score b15). Exclusion criteria included
    Potentiation between opioids and ketamine was demon-          the presence of a psychiatric history; chronic respiratory,
strated in animal studies [7] and was suggested in a              renal, or hepatic failure; known ketamine sensitivity;
volunteer study [8], whereas another report favored only          known opioid allergies; treatment of chronic pain or
an additive association [9]. Furthermore, ketamine attenu-        treatment with opioids; incapacity to understand the VAS;
ates the development of acute analgesic tolerance to opioids      pregnancy; or indication for local or regional analgesia.
in rats [10] and suppresses the rebound hyperalgesia              Patients who had already received an opioid analgesic
observed after opioid exposure in volunteers [11]. A recent       (either by self-administration or by another attending
study demonstrated that the combined administration of            physician) were also excluded.
small-dose ketamine and morphine promptly and satis-                 Intravenous morphine was given and titrated according to
factorily resolved pain that was unresponsive to intra-           the VAS pain score. Ketamine and placebo were adminis-
venous (IV) morphine alone [12]. In emergency settings, a         tered from syringes of similar appearances prepared by a
0.1-mg d kgÀ1 dose of IV morphine was not effective for           nurse anesthetist who was otherwise not involved in the
controlling severe acute pain in most patients [13]. There is     study. The dilution of ketamine was 1 mg d mLÀ1. An
evidence to suggest that the lack of effectiveness of             independent physician-observer blinded to the analgesic
morphine is due to the activation of NMDA receptors; and          treatment group did all assessments of patients. The first
if these are not effectively inhibited in time, the process may   volume administered was 0.2 mL d kgÀ1 (ie, 0.2 mg d kgÀ1)
evolve into a complex change in neural plasticity, resulting      of ketamine (Ketamine; Panpharma, France) in the K group
in central sensitization [14]. To our knowledge, there is no      or 0.2 mL d kgÀ1 of placebo in the P group given over
study assessing the interest of the morphine and ketamine         10 minutes with 0.1 mg d kgÀ1 of morphine, followed by
association for trauma patients with severe acute pain in         additional doses of 3 mg every 5 minutes until pain relief
emergency settings.                                               was obtained as defined by a VAS score not exceeding
    We tested the hypothesis that the combination of small-       30/100. The study protocol is shown in Fig. 1.
dose ketamine and morphine would promptly reduce pain                The end points of the study were the VAS and the
perception and morphine consumption compared with                 morphine consumption at 30 minutes (T30).
morphine alone in trauma patients with severe acute pain             The VAS used was a 100-mm ruler and a marker that
in emergency settings.                                            the patient moves to the point indicating his or her pain
                                                                  intensity. The VAS was presented as a horizontal line
                                                                  on which the patient’s pain intensity was represented by a
2. Materials and methods                                          point between the extremes of bno pain at allQ and bworst
                                                                  pain imaginable.Q
   We performed a prospective, multicenter, randomized,              Patients were asked to assess the intensity of their pain
double-blind, controlled study. The trial was coordinated by      by providing a VAS score on inclusion (VAS [T0]) and then
the Avicenne University Hospital (Bobigny, France). Five          every 5 minutes until arrival at hospital.
emergency departments using mobile intensive care units              All VAS scores were recorded at T0, T15, and T30.
previously described [15] were involved in this study.            Thirty minutes after the first injection, overall patients’ and
Mobile intensive care units are staffed by an attending           investigators’ satisfaction regarding analgesia (pain relief
emergency physician, a nurse anesthetist, and an emergency        classified as excellent, good, mild, or weak) was recorded.
medical technician.                                               The safety evaluation included monitoring of the blood
   The Human Subjects Committee of the Robert Ballanger           pressure, heart rate, respiratory rate, and oxygen saturation
Hospital (Aulnay, France) approved this study, and all            by pulse oximetry, as well as sedation level using the
patients provided written informed consent. Patients were         Ramsay score ranging from 1 to 6 [16] (1, anxious and
recruited between January 01, 2003, and January 31, 2005.         agitated or restless; 2, cooperative, oriented, and tranquil;
   A table of random numbers determined the randomiza-            3, responds to command only; 4, brisk response to light
tion sequence, using a restricted randomization scheme to         glabellar tap; 5, sluggish response to light glabellar tap; 6, no
ensure roughly equal numbers in each group. Group                 response to glabellar tap). Sedation was defined as a Ramsay
assignments were sealed in opaque envelopes and opened            score of greater than 2. The presence of hallucination,
sequentially by the investigators. Eligible patients were         dysphoria, weakness sensation, diplopia, nausea, vomiting,
Management of severe acute pain in emergency settings                                                                387




                         Fig. 1   The study protocol involving administration of ketamine vs placebo.


dizziness, itching, and bradypnea was likewise recorded.          score not exceeding 30/100, and standard deviation was
These data were recorded at T0 and T30.                           estimated to be approximately 15/100 [17]. To reach a
                                                                  VAS difference of more than 13/100 [18] in favor of the
2.1. Statistical analysis                                         ketamine group, the appropriate sample size using an
                                                                  a error of .05 and a b error of .10 was calculated with
   The Wilcoxon test was used for quantitative nonpara-           the formula of Casagrande et al [19]. A minimum of
metric variables such as VAS, the Student t test for              29 patients for each group should be included to see a
parametric quantitative variables, and the v 2 test for           difference of 13 mm between groups. We chose to include
qualitative variables. The aim of pain relief was a VAS           30 patients in each group to increase the power of this
388                                                                                                                        M. Galinski et al.

 Table 1      Baseline characteristics of patients from groups K
 and P
 Characteristics                        K group           P group    P
                                        (n = 33)          (n = 32)
 Age (y, mean F SD)                     35 F 13           40 F 14    .3
 Sex ratio (male-female)                25:8              23:9       .6
 BMI (kg d mÀ2, mean F SD)              25 F 4            25 F 4     .8
 Etiology of trauma (n [%])                                          .14
  Suspicion of bone fracture            19 (58)           24 (75)
  Burns                                  2 (6)             2 (6)
  Others                                12 (36)            6 (19)
 BMI indicates body mass index.

                                                                               Fig. 2 The effect of morphine with and without ketamine on the
                                                                               variation of the VAS score at T0, T15, and T30: DVAS (VAS [T0]
                                                                               À VAS [Tx]) (mean F 95% confidence interval). There was no
study. Statistical analysis was performed using Statview                       difference between the 2 groups.
Software (StatView version 5; Abacus Concepts, SAS
Institute, Berkeley, Calif). A P value of less than .05 was
considered statistically significant.                                          significantly fewer morphine boluses in the K group than
                                                                               those in the P group (Table 2).
                                                                                  The VAS score was not significantly different between
3. Results                                                                     groups K and P at T0 and T30 (Table 2). Evolution of
3.1. Patient characteristics                                                   the VAS score variation (DVAS [Tx], defined as VAS
                                                                               [T0] À AS [Tx]) was not significantly different between
   Between January 01, 2004, and June 30, 2005, seventy-                       the groups (Fig. 2). At T30, 20 patients (61%) of the
three patients were enrolled in the study (Fig. 1). Seven                      K group had a VAS score not exceeding 30/100 vs
patients (5 in the K group and 2 in the P group) were                          13 patients (41%) of the P group, representing a nonsignif-
withdrawn from the analysis because of incomplete data or                      icant difference ( P = .2).
disrespect of study protocol. One patient was excluded
because of an anaphylactoid reaction after antibiotic                          3.3. Secondary outcome
injection in the P group. Thus, data from 65 patients were                        Satisfaction of patients was not significantly different
completed and analyzed, 33 in the K group and 32 in                            between the 2 groups. There were no differences between
the P group.                                                                   groups with regard to blood pressure, heart rate, respiratory
   Baseline characteristics were similar between the                           rate, or oxygen saturation at T0 and T30 (Table 3). The
2 groups (Table 1).                                                            incidence of neuropsychological adverse effects was signif-
3.2. Primary outcome                                                           icantly greater in the K group, with hallucinations (n = 4),
                                                                               dizziness (n = 6), diplopia (n = 2), and dysphoria (n = 6),
   At T30, morphine consumption was significantly lower                        whereas there was only 1 case of dysphoria in the P group.
in the K group than that in the P group, corresponding to                      Some patients had 2 or more symptoms.



 Table 2     Comparison of morphine requirements, number of morphine boluses, VAS, and patient satisfaction between groups K and P
                                                                           K group                    P group                        P
                                                                           (n = 33)                   (n = 32)
 Morphine requirements (mg d kgÀ1, mean [95% CI])
 T0                                                                        0.099 (0.097-0.102)        0.096 (0.0917-0.100)            .2
 T30                                                                       0.149 (0.132-0.165)        0.202 (0.181-0.223)            b.001
 No. of morphine boluses (median [interquartile range])                      1.0 (0.0-2.0)               2.3 (1.8-3.8)               b.0001
 VAS (mean [95% CI])
 T0                                                                         80.4 (75.6-85.2)            80.7 (75.8-85.6)             NS
 T30                                                                        34.1 (25.6-42.6)            39.5 (32.4-46.6)             NS
 Patient satisfaction
 T30 (n [%]), excellent or good                                              18 (56)                    22 (69)                      0.3
 NS indicates not significant; CI, confidence interval.
Management of severe acute pain in emergency settings                                                                      389

 Table 3 Comparison between groups K and P for clinical
                                                                results were similar whatever their designs, namely, that
 parameters and adverse effects                                 small-dose ketamine reduced morphine requirements and
                                                                pain intensity [5,6,12]. Weinbroum [12] evaluated the
 Parameters                    K group     P group     P
                                                                effects of postoperative coadministration of small doses of
                               (n = 33)    (n = 32)
                                                                ketamine (0.250 mg d kgÀ1) and morphine on pain intensity
 Heart rate                                                     in surgical patients who complained of pain of at least 6/10
    (beats/min, mean F SD)
                                                                on a VAS despite more than 0.1 mg d kgÀ1 of IV morphine
 T0                            87 F 15     81 F 18     .3
 T30                           82 F 14     78 F 15     .5
                                                                administration within 30 minutes. This study demonstrated
 Systolic blood pressure                                        that a single dose of 0.250 mg d kgÀ1 of ketamine plus
    (mm Hg, mean F SD)                                          0.015 mg d kgÀ1 of morphine provided analgesia in 68% of
 T0                            139 F 17    139 F 21    .9       patients compared with only 3.5% of patients who received
 T30                           136 F 20    133 F 20    .5       0.030 mg d kgÀ1 of morphine with saline. Pain intensity
 Respiratory rate                                               was still lower in the former group 2 hours after.
    (breaths/min, mean F SD)                                        The idea of combining opiates and NMDA receptor
 T0                             20         19          .6       antagonists is based on numerous experimental data
 T30                            18         17          .5       suggesting different and complementary mechanisms of
 Pulse oximetry                                                 action for these 2 classes of analgesic agents [21,22].
    (%, mean F SD)
                                                                Morphine acts at both the spinal and supraspinal levels. The
 T0                             99 F 2     99 F 1      .8
 T30                            99 F 1     98 F 2      .2
                                                                spinal action is well documented [23]. It is generally
 No. of patients with           7 (21)      2 (6)      .2       accepted that opioids reduce the spinal transmission of
    Ramsay score z3 (n [%])                                     nociceptive signals predominantly at presynaptic sites
 Adverse effects (n [%])                                        (ie, reduction in transmitter release from afferent C fibers),
 Nausea and vomiting            8 (6)       4   (6)    NS       although a postsynaptic inhibition of spinal dorsal horn
 Neuropsychological            12 (36)      1   (3)    .002     nociceptive neurons has also been demonstrated. In contrast,
 Itching                        1 (3)       1   (3)    NS       NMDA receptor antagonists preferentially act postsynapti-
 Bradypnea                      0           1   (3)    NS       cally by reducing the hyperexcitability (ie, central sensiti-
                                                                zation) of spinal nociceptive neurons [21,24]. Thus, the
                                                                reduction in nociceptive inputs associated with the reduction
   The level of sedation, nausea, vomiting, and itching did     in postsynaptic neuronal hyperexcitability may explain the
not differ between the 2 groups (Table 3).                      synergistic interaction of the 2 drugs [8]. Bossard et al [8]
                                                                analyzed the effects of morphine, ketamine, and their
                                                                combination on electrophysiological recordings of the
4. Discussion                                                   nociceptive flexion RIII reflex in 12 healthy volunteers.
                                                                The stimulus response curve of the nociceptive RIII reflex
   In trauma patients with severe acute pain, we found that     was significantly reduced after injection of a combination of
the association of low-dose ketamine with morphine              ketamine and morphine, and they concluded there was a
reduced morphine requirements by approximately 26%              synergistic interaction between the 2 drugs. However, we
within 30 minutes. However, pain intensity measured on a        found that there were significantly more adverse effects
VAS at T30 was not different between the 2 groups. To our       with ketamine. Analgesic doses of 0.150 to 0.500 mg d kgÀ1
knowledge, this is the first study that associated morphine     of ketamine have been reported to produce dose- or plasma
with low-dose ketamine for trauma patients with severe          concentration–dependent cognitive, perceptual, and mood
acute pain in emergency settings.                               disturbances, as well as psychotomimetic adverse effects
   Gurnani et al [20] studied low-dose ketamine in patients     [25,26]. This could be a limitation to the use of ketamine in
with acute pain after musculoskeletal trauma in emergency       such settings, especially if the benefit regarding pain is poor.
settings. However, in this study, patients initially received   In our study, all adverse effects were weak and brief because
0.250 mg d kgÀ1 of IV ketamine, followed by a constant-rate     no one needed treatment and patients’ satisfaction was not
infusion of subcutaneous ketamine (0.100 mg d kgÀ1 d hÀ1)       different between the 2 groups. In a review article about
without morphine, whereas the control group received IV         perioperative ketamine for acute postoperative pain, Bell
injections of morphine (0.1 mg d kgÀ1 every 4 hours). They      et al [27] found that the occurrence of adverse effects was
found that the onset of analgesia was slower in the morphine    similar in ketamine- and placebo-treated groups. Especially,
group and that analgesia was more intense in the ketamine       21 of 37 trials specifically stated that there were no
group [20]. Furthermore, the rate of patients requiring         psychotomimetic adverse effects. However, in an experi-
supplementary analgesia was lower in the ketamine group.        mental study on 12 healthy volunteers, Bossard et al found
However, these patients did not have severe pain.               many psychosensory effects (8-10/12 volunteers) after the
   Most studies about small doses of ketamine in painful        administration of ketamine, morphine, or their combination,
patients were performed in postoperative settings. Their        but the incidence was similar between groups.
390                                                                                                                               M. Galinski et al.

   The aim of a coanalgesic association is to avoid opioid                      [9] Sethna NF, Liu M, Gracely R, et al. Analgesic and cognitive effects of
overdose and adverse effects with equal or better analgesia.                        intravenous ketamine-alfentanil combinations versus either drug alone
                                                                                    after intradermal capsaicin in normal subjects. Anesth Analg 1998;
Weinbroum [12] obtained significantly fewer adverse effects                         86:1250 - 6.
such as nausea and vomiting with ketamine and morphine                         [10] Kissin I, Bright CA, Bradley Jr EL. The effect of ketamine on opioid-
compared with morphine and placebo (12% and 38%,                                    induced acute tolerance: can it explain reduction of opioid consump-
respectively). We did not reach this conclusion because it                          tion with ketamine-opioid analgesic combinations? Anesth Analg
was not the first end point of our study and the patient                            2000;91:1483 - 8.
                                                                               [11] Angst MS, Koppert W, Pahl I, et al. Short-term infusion of the A-
number was too small for that.
                                                                                    opioid agonist remifentanil in humans causes hyperalgesia during
   The observation period also has to be longer than                                withdrawal. Pain 2003;106:49 - 57.
30 minutes because there is probably a ketamine activity                       [12] Weinbroum AA. A single small dose of postoperative ketamine
that lasts longer than its simple pharmacokinetic action                            provides rapid and sustained improvement in morphine analgesia
particularly on hyperalgesia. This point was demonstrated in                        in the presence of morphine-resistant pain. Anesth Analg 2003;
postoperative studies [12]. Furthermore, the ketamine we                            96:789 - 95.
                                                                               [13] Bijur PE, Kenny MK, Gallagher EJ. Intravenous morphine at
used was a racemic mixture consisting of equal shares of                            0.1 mg/kg is not effective for controlling severe acute pain in the
2 optical enantiomers. Pharmacologic investigations show                            majority of patients. Ann Emerg Med 2005;46:362 - 5.
differences between those enantiomers, with a clinical                         [14] Mao J, Price DD, Mayer DJ. Mechanisms of hyperalgesia and
superiority of S(+)-ketamine particularly with fewer adverse                        morphine tolerance: a current view of their possible interaction. Pain
effects [28]. Unfortunately, this enantiomer is not yet                             1995;62:259 - 74.
                                                                               [15] Adnet F, Jouriles NJ, Le Toumelin P, Hennequin B, Taillandier C,
available in our country.
                                                                                    Rayeh F, et al. Survey of out-of-hospital emergency intubations in the
   The observation period of our trial is a limitation because                      French prehospital medical system: a multicenter study. Ann Emerg
30 minutes was probably too short. Indeed, Weinbroum [12]                           Med 1998;32:454 - 60.
demonstrated that the analgesic effects of the ketamine                        [16] Ramsay MA, Savege TM, Simpson BR, Gooodwin R. Controlled
group were clearly evident throughout the 120-minute                                sedation with alphaxalone-alphadolone. Br J Med 1974;2:256 - 9.
                                                                               [17] Ricard-Hibon A, Chollet C, Saada S, et al. A quality control program
observation period.
                                                                                    for acute pain management in out-of-hospital critical care medicine.
                                                                                    Ann Emerg Med 1999;34:738 - 44.
5. Conclusion                                                                  [18] Todd KH, Funk KG, Funk JP, Bonacci R. Clinical significance
                                                                                    of reported changes in pain severity. Ann Emerg Med 1996;27:
   This study showed that low-dose ketamine in trauma                               485 - 9.
                                                                               [19] Casagrande JT, Pike MC, Smith PG. An improved approximate
patients with severe acute pain reduced morphine require-                           formula for calculating sample sizes for comparing two binomial
ments. We could not demonstrate any reduction in pain                               distributions. Biometrics 1978;34:483 - 6.
intensity with ketamine in this study.                                         [20] Gurnani A, Sharma PK, Rautela RS, Bhattacharya A. Analgesia for
                                                                                    acute musculoskeletal trauma: low-dose subcutaneous infusion of
                                                                                    ketamine. Anaesth Intensive Care 1996;24:32 - 6.
References                                                                     [21] Dickenson AH. NMDA receptor antagonists: interactions with
                                                                                    opioids. Acta Anaesthesiol Scand 1997;41:112 - 5.
 [1] Belpomme V, Ricard-Hibon A, Cholet C, et al. Delai d’obtention de
                                                        ´                      [22] Wiesenfeld-Hallin Z. Combined opioid-NMDA antagonist therapies.
     l’analgesie en prehospitalier. Ann Fr Anesth Reanim 2002;21:324S
             ´         ´                                                            What advantages do they offer the control of pain syndromes? Drugs
     [abstract].                                                                    1998;56:1 - 5.
 [2] Paqueron X, Lumbroso A, Mergoni P, et al. Is morphine-induced             [23] Yaksh TL. Pharmacology and mechanisms of opioid analgesic
     sedation synonymous with analgesia during intravenous morphine                 activity. Acta Anaesthesiol Scand 1997;41:94 - 111.
     titration? Br J Anaesth 2002;89:697 - 701.                                [24] Chapman V, Dickenson AH. The combination of NMDA antagonism
 [3] Kehlet H, Dahl JB. The value of bmultimodalQ or bbalanced analgesiaQ           and morphine produces profound antinociception in the rat dorsal
     in postoperative pain treatment. Anesth-Analg 1993;77:1048 - 56.               horn. Brain Res 1992;573:321 - 3.
 [4] Richebe P, Rivat C, Rivalan B, Maurette P, Simonnet G. Low doses
              ´                                                                [25] Oye I, Paulsen O, Maurset A. Effects of ketamine on sensory
     ketamine: antihyperalgesic drug, non-analgesic. Ann Fr Anesth                  perception: evidence for a role of N-methyl-d-aspartate receptors.
     Reanim 2005;24:11349 - 59.                                                     J Pharmacol Exp Ther 1992;260:1209 - 13.
 [5] Kapfer B, Alfonsi P, Guignard B, Sessler DI, Chauvin M. Nefopam           [26] Krystal JH, Karper LP, Seibyl JP, et al. Subanesthetic effects of
     and ketamine comparably enhance postoperative analgesia. Anesth                noncompetitive NMDA antagonist, ketamine, in humans: psychomi-
     Analg 2005;100:169 - 74.                                                       metic, perceptual, cognitive and neuroendocrine responses. Arch Gen
 [6] Suzuki M, Kentaro T, Lansing PS, Tolan MM, Fuhrman TM, Ignacio                 Psychiatry 1994;14:144 - 53.
     CI, et al. Small-dose ketamine enhances morphine-induced analgesia        [27] Bell RF, Dahl JB, Moore RA, Kalso E. Perioperative ketamine
     after outpatient surgery. Anesth Analg 1999;89:98 - 103.                       for acute post-operative pain: a quantitative and qualitative systema-
 [7] Alvarez P, Saavedra G, Hernandez A, et al. Synergistic antinociceptive         tic review (Cochrane review). Acta Anaesthesiol Scand 2005;49:
     effects of ketamine and morphine in the orofacial capsaicin test in the        1405 - 28.
     rat. Anesthesiology 2003;99:969 - 75.                                     [28] Pfenninger EG, Durieux ME, Himmelseher S. Cognitive impairment
 [8] Bossard AE, Guirimand F, Fletcher D, et al. Interaction of a                   after small-dose ketamine isomer in comparison to equianalgesic
     combination of morphine and ketamine on the nociceptive flexion                racemic ketamine in human volunteers. Anesthesiology 2002;
     reflex in human volunteers. Pain 2002;98:47 - 57.                              96:357 - 66.

More Related Content

What's hot

1946 Pain in men wounded in battle
1946 Pain in men wounded in battle 1946 Pain in men wounded in battle
1946 Pain in men wounded in battle Robert Cole
 
A prospective, randomized, double blind study to evaluate Morphine sparing ef...
A prospective, randomized, double blind study to evaluate Morphine sparing ef...A prospective, randomized, double blind study to evaluate Morphine sparing ef...
A prospective, randomized, double blind study to evaluate Morphine sparing ef...iosrjce
 
CHRONIC PAIN AND DEPRESSION: Cause or Effect or Linked?
CHRONIC PAIN AND DEPRESSION: Cause or Effect or Linked?CHRONIC PAIN AND DEPRESSION: Cause or Effect or Linked?
CHRONIC PAIN AND DEPRESSION: Cause or Effect or Linked?Sudhir Kumar
 
Second life of Ketamine
Second life of KetamineSecond life of Ketamine
Second life of Ketaminemjsigtermans
 
Pain Therapeutics 2017
Pain Therapeutics 2017Pain Therapeutics 2017
Pain Therapeutics 2017Teri Arri
 
Pain Management Current & Newer Modalities
Pain Management Current & Newer Modalities Pain Management Current & Newer Modalities
Pain Management Current & Newer Modalities Dr Sachin Pawar
 
Risk-Benefit High-Dose LTOT
Risk-Benefit High-Dose LTOTRisk-Benefit High-Dose LTOT
Risk-Benefit High-Dose LTOTPaul Coelho, MD
 
Per-operative Pain Managment in Children
Per-operative Pain Managment in ChildrenPer-operative Pain Managment in Children
Per-operative Pain Managment in ChildrenDr.Mahmoud Abbas
 
Topical & Transdermal Medications in Palliative Medicine
Topical & Transdermal Medications in Palliative MedicineTopical & Transdermal Medications in Palliative Medicine
Topical & Transdermal Medications in Palliative MedicineChristian Sinclair
 
Pain control in Emergency Department
Pain control in Emergency DepartmentPain control in Emergency Department
Pain control in Emergency DepartmentDr.Mahmoud Abbas
 
Treating chronic pain with cannabis-derived compounds
Treating chronic pain with cannabis-derived compoundsTreating chronic pain with cannabis-derived compounds
Treating chronic pain with cannabis-derived compoundsAbdul Rehman Mohammad
 
Topical paincontrolmedication
Topical paincontrolmedicationTopical paincontrolmedication
Topical paincontrolmedicationpiushjpatel
 
Medication administration errors associated with transitions of care in inpat...
Medication administration errors associated with transitions of care in inpat...Medication administration errors associated with transitions of care in inpat...
Medication administration errors associated with transitions of care in inpat...Canadian Patient Safety Institute
 
An overview of drug regulatory system in South Africa
An overview of drug regulatory system in South AfricaAn overview of drug regulatory system in South Africa
An overview of drug regulatory system in South AfricaSriramNagarajan17
 
Compounding for Hospice Patients
Compounding for Hospice PatientsCompounding for Hospice Patients
Compounding for Hospice Patientsplacenc
 
Habituation letter to editor (1)
Habituation letter to editor (1)Habituation letter to editor (1)
Habituation letter to editor (1)Nchedo Okafor
 
Wright-Williams et al (2013) Effects of vasectomy & buprenorphine on cort and...
Wright-Williams et al (2013) Effects of vasectomy & buprenorphine on cort and...Wright-Williams et al (2013) Effects of vasectomy & buprenorphine on cort and...
Wright-Williams et al (2013) Effects of vasectomy & buprenorphine on cort and...Johnny Roughan
 

What's hot (20)

1946 Pain in men wounded in battle
1946 Pain in men wounded in battle 1946 Pain in men wounded in battle
1946 Pain in men wounded in battle
 
A prospective, randomized, double blind study to evaluate Morphine sparing ef...
A prospective, randomized, double blind study to evaluate Morphine sparing ef...A prospective, randomized, double blind study to evaluate Morphine sparing ef...
A prospective, randomized, double blind study to evaluate Morphine sparing ef...
 
CHRONIC PAIN AND DEPRESSION: Cause or Effect or Linked?
CHRONIC PAIN AND DEPRESSION: Cause or Effect or Linked?CHRONIC PAIN AND DEPRESSION: Cause or Effect or Linked?
CHRONIC PAIN AND DEPRESSION: Cause or Effect or Linked?
 
Second life of Ketamine
Second life of KetamineSecond life of Ketamine
Second life of Ketamine
 
Pain Therapeutics 2017
Pain Therapeutics 2017Pain Therapeutics 2017
Pain Therapeutics 2017
 
Pain Management Current & Newer Modalities
Pain Management Current & Newer Modalities Pain Management Current & Newer Modalities
Pain Management Current & Newer Modalities
 
Risk-Benefit High-Dose LTOT
Risk-Benefit High-Dose LTOTRisk-Benefit High-Dose LTOT
Risk-Benefit High-Dose LTOT
 
Per-operative Pain Managment in Children
Per-operative Pain Managment in ChildrenPer-operative Pain Managment in Children
Per-operative Pain Managment in Children
 
Topical & Transdermal Medications in Palliative Medicine
Topical & Transdermal Medications in Palliative MedicineTopical & Transdermal Medications in Palliative Medicine
Topical & Transdermal Medications in Palliative Medicine
 
Qysmia
QysmiaQysmia
Qysmia
 
Pain control in Emergency Department
Pain control in Emergency DepartmentPain control in Emergency Department
Pain control in Emergency Department
 
Treating chronic pain with cannabis-derived compounds
Treating chronic pain with cannabis-derived compoundsTreating chronic pain with cannabis-derived compounds
Treating chronic pain with cannabis-derived compounds
 
Topical paincontrolmedication
Topical paincontrolmedicationTopical paincontrolmedication
Topical paincontrolmedication
 
Cancer pain
Cancer painCancer pain
Cancer pain
 
Medication administration errors associated with transitions of care in inpat...
Medication administration errors associated with transitions of care in inpat...Medication administration errors associated with transitions of care in inpat...
Medication administration errors associated with transitions of care in inpat...
 
An overview of drug regulatory system in South Africa
An overview of drug regulatory system in South AfricaAn overview of drug regulatory system in South Africa
An overview of drug regulatory system in South Africa
 
Compounding for Hospice Patients
Compounding for Hospice PatientsCompounding for Hospice Patients
Compounding for Hospice Patients
 
Multimodal Regiments for Acute Pain Management - Prof. A. Husni Tanra
Multimodal Regiments for Acute  Pain Management - Prof. A. Husni TanraMultimodal Regiments for Acute  Pain Management - Prof. A. Husni Tanra
Multimodal Regiments for Acute Pain Management - Prof. A. Husni Tanra
 
Habituation letter to editor (1)
Habituation letter to editor (1)Habituation letter to editor (1)
Habituation letter to editor (1)
 
Wright-Williams et al (2013) Effects of vasectomy & buprenorphine on cort and...
Wright-Williams et al (2013) Effects of vasectomy & buprenorphine on cort and...Wright-Williams et al (2013) Effects of vasectomy & buprenorphine on cort and...
Wright-Williams et al (2013) Effects of vasectomy & buprenorphine on cort and...
 

Similar to Management Of Severe Acute Pain In Emergency Settings Ketamine Reduces Morphine Consumption

Marineo + smith jan 2012 scrambler therapy better than drugs marineo 2012
Marineo + smith jan 2012 scrambler therapy better than drugs  marineo 2012Marineo + smith jan 2012 scrambler therapy better than drugs  marineo 2012
Marineo + smith jan 2012 scrambler therapy better than drugs marineo 2012Calmar Pain Relief Therapy, LLC
 
ORIGINAL CONTRIBUTIONPatient-Controlled Transdermal Fentan.docx
ORIGINAL CONTRIBUTIONPatient-Controlled Transdermal Fentan.docxORIGINAL CONTRIBUTIONPatient-Controlled Transdermal Fentan.docx
ORIGINAL CONTRIBUTIONPatient-Controlled Transdermal Fentan.docxgerardkortney
 
Journal Club Trigeminal Neuralgia
Journal Club Trigeminal NeuralgiaJournal Club Trigeminal Neuralgia
Journal Club Trigeminal NeuralgiaArun Panwar
 
Keta a ic en mastectomía.pdf
Keta a ic en mastectomía.pdfKeta a ic en mastectomía.pdf
Keta a ic en mastectomía.pdfleroleroero1
 
Physician-Pharmacist Comanagement of Postoperative Pain in Egyptian Patients:...
Physician-Pharmacist Comanagement of Postoperative Pain in Egyptian Patients:...Physician-Pharmacist Comanagement of Postoperative Pain in Egyptian Patients:...
Physician-Pharmacist Comanagement of Postoperative Pain in Egyptian Patients:...iosrphr_editor
 
Pharmacist Educational Intervention in Intravenous Patient Controlled Analges...
Pharmacist Educational Intervention in Intravenous Patient Controlled Analges...Pharmacist Educational Intervention in Intravenous Patient Controlled Analges...
Pharmacist Educational Intervention in Intravenous Patient Controlled Analges...Sunil Vadithya
 
Chloroprocaine study
Chloroprocaine studyChloroprocaine study
Chloroprocaine studyVenesa Ingold
 
buprenorfina y medetomidina en gatos.pdf
buprenorfina y medetomidina en gatos.pdfbuprenorfina y medetomidina en gatos.pdf
buprenorfina y medetomidina en gatos.pdfleroleroero1
 
PONV_VMoll1_2012.pptx
PONV_VMoll1_2012.pptxPONV_VMoll1_2012.pptx
PONV_VMoll1_2012.pptxFiNi17
 
Integrated High-Risk Pain Management Clinic: Addressing co-morbid pain and de...
Integrated High-Risk Pain Management Clinic: Addressing co-morbid pain and de...Integrated High-Risk Pain Management Clinic: Addressing co-morbid pain and de...
Integrated High-Risk Pain Management Clinic: Addressing co-morbid pain and de...Michael Changaris
 
Post herpetic neuralgia
Post herpetic neuralgiaPost herpetic neuralgia
Post herpetic neuralgiayury
 
Pain Relief In Labour1
Pain  Relief In  Labour1Pain  Relief In  Labour1
Pain Relief In Labour1inojustin
 
Clonidine as an_adjuvant_to_local_anesthetics_for.28
Clonidine as an_adjuvant_to_local_anesthetics_for.28Clonidine as an_adjuvant_to_local_anesthetics_for.28
Clonidine as an_adjuvant_to_local_anesthetics_for.28Telmo Santos
 
Interventional Techniques For Cancer Pain Management.
Interventional Techniques For Cancer Pain Management.Interventional Techniques For Cancer Pain Management.
Interventional Techniques For Cancer Pain Management.guest7342323
 
Vaso-occlusive Crisis.pptx
Vaso-occlusive Crisis.pptxVaso-occlusive Crisis.pptx
Vaso-occlusive Crisis.pptxChristineiyke
 

Similar to Management Of Severe Acute Pain In Emergency Settings Ketamine Reduces Morphine Consumption (20)

Marineo + smith jan 2012 scrambler therapy better than drugs marineo 2012
Marineo + smith jan 2012 scrambler therapy better than drugs  marineo 2012Marineo + smith jan 2012 scrambler therapy better than drugs  marineo 2012
Marineo + smith jan 2012 scrambler therapy better than drugs marineo 2012
 
ORIGINAL CONTRIBUTIONPatient-Controlled Transdermal Fentan.docx
ORIGINAL CONTRIBUTIONPatient-Controlled Transdermal Fentan.docxORIGINAL CONTRIBUTIONPatient-Controlled Transdermal Fentan.docx
ORIGINAL CONTRIBUTIONPatient-Controlled Transdermal Fentan.docx
 
Journal Club Trigeminal Neuralgia
Journal Club Trigeminal NeuralgiaJournal Club Trigeminal Neuralgia
Journal Club Trigeminal Neuralgia
 
Keta a ic en mastectomía.pdf
Keta a ic en mastectomía.pdfKeta a ic en mastectomía.pdf
Keta a ic en mastectomía.pdf
 
Physician-Pharmacist Comanagement of Postoperative Pain in Egyptian Patients:...
Physician-Pharmacist Comanagement of Postoperative Pain in Egyptian Patients:...Physician-Pharmacist Comanagement of Postoperative Pain in Egyptian Patients:...
Physician-Pharmacist Comanagement of Postoperative Pain in Egyptian Patients:...
 
tramadol
tramadoltramadol
tramadol
 
Fentanyl for perioperative pain management - Dr. Alex Yeo Sow Nam
Fentanyl for perioperative pain management - Dr. Alex Yeo Sow NamFentanyl for perioperative pain management - Dr. Alex Yeo Sow Nam
Fentanyl for perioperative pain management - Dr. Alex Yeo Sow Nam
 
Pharmacist Educational Intervention in Intravenous Patient Controlled Analges...
Pharmacist Educational Intervention in Intravenous Patient Controlled Analges...Pharmacist Educational Intervention in Intravenous Patient Controlled Analges...
Pharmacist Educational Intervention in Intravenous Patient Controlled Analges...
 
Chloroprocaine study
Chloroprocaine studyChloroprocaine study
Chloroprocaine study
 
buprenorfina y medetomidina en gatos.pdf
buprenorfina y medetomidina en gatos.pdfbuprenorfina y medetomidina en gatos.pdf
buprenorfina y medetomidina en gatos.pdf
 
PONV_VMoll1_2012.pptx
PONV_VMoll1_2012.pptxPONV_VMoll1_2012.pptx
PONV_VMoll1_2012.pptx
 
remifentanil
remifentanilremifentanil
remifentanil
 
KETAMINE-5.ppt
KETAMINE-5.pptKETAMINE-5.ppt
KETAMINE-5.ppt
 
Integrated High-Risk Pain Management Clinic: Addressing co-morbid pain and de...
Integrated High-Risk Pain Management Clinic: Addressing co-morbid pain and de...Integrated High-Risk Pain Management Clinic: Addressing co-morbid pain and de...
Integrated High-Risk Pain Management Clinic: Addressing co-morbid pain and de...
 
postop analgesia.pdf
postop analgesia.pdfpostop analgesia.pdf
postop analgesia.pdf
 
Post herpetic neuralgia
Post herpetic neuralgiaPost herpetic neuralgia
Post herpetic neuralgia
 
Pain Relief In Labour1
Pain  Relief In  Labour1Pain  Relief In  Labour1
Pain Relief In Labour1
 
Clonidine as an_adjuvant_to_local_anesthetics_for.28
Clonidine as an_adjuvant_to_local_anesthetics_for.28Clonidine as an_adjuvant_to_local_anesthetics_for.28
Clonidine as an_adjuvant_to_local_anesthetics_for.28
 
Interventional Techniques For Cancer Pain Management.
Interventional Techniques For Cancer Pain Management.Interventional Techniques For Cancer Pain Management.
Interventional Techniques For Cancer Pain Management.
 
Vaso-occlusive Crisis.pptx
Vaso-occlusive Crisis.pptxVaso-occlusive Crisis.pptx
Vaso-occlusive Crisis.pptx
 

More from EM OMSB

Case presentation
Case presentationCase presentation
Case presentationEM OMSB
 
Heroic procedures you should know
Heroic procedures you should knowHeroic procedures you should know
Heroic procedures you should knowEM OMSB
 
Ed overcrowding
Ed overcrowdingEd overcrowding
Ed overcrowdingEM OMSB
 
challenge rash
 challenge rash challenge rash
challenge rashEM OMSB
 
Case Presenation
Case PresenationCase Presenation
Case PresenationEM OMSB
 
Clinical Series Pesticide
Clinical Series PesticideClinical Series Pesticide
Clinical Series PesticideEM OMSB
 
The seizing patient
The seizing patientThe seizing patient
The seizing patientEM OMSB
 
Coccain and Sympathomimatic
Coccain and Sympathomimatic Coccain and Sympathomimatic
Coccain and Sympathomimatic EM OMSB
 
Case presentation
Case presentationCase presentation
Case presentationEM OMSB
 
Venomous marine
Venomous marineVenomous marine
Venomous marineEM OMSB
 
Optimzing sepsis management
Optimzing sepsis managementOptimzing sepsis management
Optimzing sepsis managementEM OMSB
 
Heavy metals iron and lithium
Heavy metals iron and lithiumHeavy metals iron and lithium
Heavy metals iron and lithiumEM OMSB
 
Antibiotic in ED
Antibiotic in EDAntibiotic in ED
Antibiotic in EDEM OMSB
 
Aortic disasters ahmed
Aortic disasters ahmedAortic disasters ahmed
Aortic disasters ahmedEM OMSB
 
Case Presentation
Case Presentation Case Presentation
Case Presentation EM OMSB
 
Clinical emergency procedures Chest Tube
Clinical emergency procedures Chest TubeClinical emergency procedures Chest Tube
Clinical emergency procedures Chest TubeEM OMSB
 
Resuscitation in special populations
Resuscitation in special populationsResuscitation in special populations
Resuscitation in special populationsEM OMSB
 
NIV updated
NIV updatedNIV updated
NIV updatedEM OMSB
 
RAA SEPT 7TH
RAA SEPT 7THRAA SEPT 7TH
RAA SEPT 7THEM OMSB
 
Raa blog
Raa blogRaa blog
Raa blogEM OMSB
 

More from EM OMSB (20)

Case presentation
Case presentationCase presentation
Case presentation
 
Heroic procedures you should know
Heroic procedures you should knowHeroic procedures you should know
Heroic procedures you should know
 
Ed overcrowding
Ed overcrowdingEd overcrowding
Ed overcrowding
 
challenge rash
 challenge rash challenge rash
challenge rash
 
Case Presenation
Case PresenationCase Presenation
Case Presenation
 
Clinical Series Pesticide
Clinical Series PesticideClinical Series Pesticide
Clinical Series Pesticide
 
The seizing patient
The seizing patientThe seizing patient
The seizing patient
 
Coccain and Sympathomimatic
Coccain and Sympathomimatic Coccain and Sympathomimatic
Coccain and Sympathomimatic
 
Case presentation
Case presentationCase presentation
Case presentation
 
Venomous marine
Venomous marineVenomous marine
Venomous marine
 
Optimzing sepsis management
Optimzing sepsis managementOptimzing sepsis management
Optimzing sepsis management
 
Heavy metals iron and lithium
Heavy metals iron and lithiumHeavy metals iron and lithium
Heavy metals iron and lithium
 
Antibiotic in ED
Antibiotic in EDAntibiotic in ED
Antibiotic in ED
 
Aortic disasters ahmed
Aortic disasters ahmedAortic disasters ahmed
Aortic disasters ahmed
 
Case Presentation
Case Presentation Case Presentation
Case Presentation
 
Clinical emergency procedures Chest Tube
Clinical emergency procedures Chest TubeClinical emergency procedures Chest Tube
Clinical emergency procedures Chest Tube
 
Resuscitation in special populations
Resuscitation in special populationsResuscitation in special populations
Resuscitation in special populations
 
NIV updated
NIV updatedNIV updated
NIV updated
 
RAA SEPT 7TH
RAA SEPT 7THRAA SEPT 7TH
RAA SEPT 7TH
 
Raa blog
Raa blogRaa blog
Raa blog
 

Management Of Severe Acute Pain In Emergency Settings Ketamine Reduces Morphine Consumption

  • 1. American Journal of Emergency Medicine (2007) 25, 385 – 390 www.elsevier.com/locate/ajem Original Contribution Management of severe acute pain in emergency settings: ketamine reduces morphine consumptionB Michel Galinski MDa,*, Francois Dolveck MDb, Xavier Combes MDe, ¸ Veronique Limoges MD , Nadia Smail MD, PhDd, Veronique Pommier MDc, ´ c !! ´ b a Francois Templier MD , Jean Catineau MD , ¸ Frederic Lapostolle MDa, Frederic Adnet MD, PhDa ´ ´ ´ ´ a Samu 93-EA 3409 Avicenne Hospital, 93009 Bobigny Cedex, France b Samu 92-Raymond Poincare Hospital, 92100 Garches, France ´ c Emergency Department-Smur 77, Meaux’s Hospital, 77100 Meaux, France d Samu 31-Purpan Hospital, 31000 Toulouses, France e ´ Samu 94-Henri Mondor Hospital, 94000 Creteil, France Received 1 July 2006; revised 1 November 2006; accepted 2 November 2006 Abstract Objective: The aim of the study was to compare in emergency settings 2 analgesic regimens, morphine with ketamine (K group) or morphine with placebo (P group), for severe acute pain in trauma patients. Methods: This was a prospective, multicenter, randomized, double-blind, clinical trial. Seventy-three trauma patients with a severe acute pain defined as a visual analog scale (VAS) score of at least 60/100 were enrolled. Patients in the K group received 0.2 mg d kgÀ1 of intravenous ketamine over 10 minutes, and patients in the P group received isotonic sodium chloride solution. In both groups, patients were given an initial intravenous morphine injection of 0.1 mg d kgÀ1, followed by 3 mg every 5 minutes. Efficient analgesia was defined as a VAS score not exceeding 30/100. The primary end points were morphine consumption and VAS at 30 minutes (T30). Results: At T30, morphine consumption was significantly lower in the K group vs the P group, with 0.149 mg d kgÀ1 (0.132-0.165) and 0.202 mg d kgÀ1 (0.181-0.223), respectively ( P b .001). The VAS score at T30 did not differ significantly between the 2 groups, with 34.1 (25.6-42.6) in the K group and 39.5 (32.4-46.6) in the P group ( P = not significant). Conclusion: Ketamine was able to provide a morphine-sparing effect. D 2007 Elsevier Inc. All rights reserved. 1. Introduction Presented at the European Society of Anaesthesiology Congress in Madrid, Spain, June 3-6, 2006. B Morphine titration infusion usually provides rapid and Support was provided solely by institutional and/or departmental sources. effective analgesia in severe acute pain [1]. However, * Corresponding author. Tel.: +33 1 48 96 44 55; fax: +33 1 48 96 44 45. adverse effects sometimes occur and may require discon- E-mail address: michel.galinski@avc.ap-hop-paris.fr (M. Galinski). tinuation of morphine titration before sufficient pain relief is 0735-6757/$ – see front matter D 2007 Elsevier Inc. All rights reserved. doi:10.1016/j.ajem.2006.11.016
  • 2. 386 M. Galinski et al. obtained [2]. The combination of nonopioid analgesics with randomly allocated to receive either morphine and ket- morphine provides a morphine-sparing effect and should amine (K group) or morphine and placebo (P group). decrease toxicity. This concept is the basis of multimodal Patients were eligible for inclusion if they presented a analgesia [3]. trauma with a severe acute pain defined as a visual analog Small doses of ketamine possess N-methyl-d-aspartate scale (VAS) score of at least 60/100; were aged between (NMDA) receptor noncompetitive antagonist properties 18 and 70 years; and were without acute respiratory, through a magnesium-dependent channel blockade [4]. hemodynamic, or neurologic compromise (respiratory Several studies demonstrated that it improves opioid distress signs, systolic blood pressure V90 mm Hg, analgesia in postoperative settings [5,6]. Glasgow Coma Score b15). Exclusion criteria included Potentiation between opioids and ketamine was demon- the presence of a psychiatric history; chronic respiratory, strated in animal studies [7] and was suggested in a renal, or hepatic failure; known ketamine sensitivity; volunteer study [8], whereas another report favored only known opioid allergies; treatment of chronic pain or an additive association [9]. Furthermore, ketamine attenu- treatment with opioids; incapacity to understand the VAS; ates the development of acute analgesic tolerance to opioids pregnancy; or indication for local or regional analgesia. in rats [10] and suppresses the rebound hyperalgesia Patients who had already received an opioid analgesic observed after opioid exposure in volunteers [11]. A recent (either by self-administration or by another attending study demonstrated that the combined administration of physician) were also excluded. small-dose ketamine and morphine promptly and satis- Intravenous morphine was given and titrated according to factorily resolved pain that was unresponsive to intra- the VAS pain score. Ketamine and placebo were adminis- venous (IV) morphine alone [12]. In emergency settings, a tered from syringes of similar appearances prepared by a 0.1-mg d kgÀ1 dose of IV morphine was not effective for nurse anesthetist who was otherwise not involved in the controlling severe acute pain in most patients [13]. There is study. The dilution of ketamine was 1 mg d mLÀ1. An evidence to suggest that the lack of effectiveness of independent physician-observer blinded to the analgesic morphine is due to the activation of NMDA receptors; and treatment group did all assessments of patients. The first if these are not effectively inhibited in time, the process may volume administered was 0.2 mL d kgÀ1 (ie, 0.2 mg d kgÀ1) evolve into a complex change in neural plasticity, resulting of ketamine (Ketamine; Panpharma, France) in the K group in central sensitization [14]. To our knowledge, there is no or 0.2 mL d kgÀ1 of placebo in the P group given over study assessing the interest of the morphine and ketamine 10 minutes with 0.1 mg d kgÀ1 of morphine, followed by association for trauma patients with severe acute pain in additional doses of 3 mg every 5 minutes until pain relief emergency settings. was obtained as defined by a VAS score not exceeding We tested the hypothesis that the combination of small- 30/100. The study protocol is shown in Fig. 1. dose ketamine and morphine would promptly reduce pain The end points of the study were the VAS and the perception and morphine consumption compared with morphine consumption at 30 minutes (T30). morphine alone in trauma patients with severe acute pain The VAS used was a 100-mm ruler and a marker that in emergency settings. the patient moves to the point indicating his or her pain intensity. The VAS was presented as a horizontal line on which the patient’s pain intensity was represented by a 2. Materials and methods point between the extremes of bno pain at allQ and bworst pain imaginable.Q We performed a prospective, multicenter, randomized, Patients were asked to assess the intensity of their pain double-blind, controlled study. The trial was coordinated by by providing a VAS score on inclusion (VAS [T0]) and then the Avicenne University Hospital (Bobigny, France). Five every 5 minutes until arrival at hospital. emergency departments using mobile intensive care units All VAS scores were recorded at T0, T15, and T30. previously described [15] were involved in this study. Thirty minutes after the first injection, overall patients’ and Mobile intensive care units are staffed by an attending investigators’ satisfaction regarding analgesia (pain relief emergency physician, a nurse anesthetist, and an emergency classified as excellent, good, mild, or weak) was recorded. medical technician. The safety evaluation included monitoring of the blood The Human Subjects Committee of the Robert Ballanger pressure, heart rate, respiratory rate, and oxygen saturation Hospital (Aulnay, France) approved this study, and all by pulse oximetry, as well as sedation level using the patients provided written informed consent. Patients were Ramsay score ranging from 1 to 6 [16] (1, anxious and recruited between January 01, 2003, and January 31, 2005. agitated or restless; 2, cooperative, oriented, and tranquil; A table of random numbers determined the randomiza- 3, responds to command only; 4, brisk response to light tion sequence, using a restricted randomization scheme to glabellar tap; 5, sluggish response to light glabellar tap; 6, no ensure roughly equal numbers in each group. Group response to glabellar tap). Sedation was defined as a Ramsay assignments were sealed in opaque envelopes and opened score of greater than 2. The presence of hallucination, sequentially by the investigators. Eligible patients were dysphoria, weakness sensation, diplopia, nausea, vomiting,
  • 3. Management of severe acute pain in emergency settings 387 Fig. 1 The study protocol involving administration of ketamine vs placebo. dizziness, itching, and bradypnea was likewise recorded. score not exceeding 30/100, and standard deviation was These data were recorded at T0 and T30. estimated to be approximately 15/100 [17]. To reach a VAS difference of more than 13/100 [18] in favor of the 2.1. Statistical analysis ketamine group, the appropriate sample size using an a error of .05 and a b error of .10 was calculated with The Wilcoxon test was used for quantitative nonpara- the formula of Casagrande et al [19]. A minimum of metric variables such as VAS, the Student t test for 29 patients for each group should be included to see a parametric quantitative variables, and the v 2 test for difference of 13 mm between groups. We chose to include qualitative variables. The aim of pain relief was a VAS 30 patients in each group to increase the power of this
  • 4. 388 M. Galinski et al. Table 1 Baseline characteristics of patients from groups K and P Characteristics K group P group P (n = 33) (n = 32) Age (y, mean F SD) 35 F 13 40 F 14 .3 Sex ratio (male-female) 25:8 23:9 .6 BMI (kg d mÀ2, mean F SD) 25 F 4 25 F 4 .8 Etiology of trauma (n [%]) .14 Suspicion of bone fracture 19 (58) 24 (75) Burns 2 (6) 2 (6) Others 12 (36) 6 (19) BMI indicates body mass index. Fig. 2 The effect of morphine with and without ketamine on the variation of the VAS score at T0, T15, and T30: DVAS (VAS [T0] À VAS [Tx]) (mean F 95% confidence interval). There was no study. Statistical analysis was performed using Statview difference between the 2 groups. Software (StatView version 5; Abacus Concepts, SAS Institute, Berkeley, Calif). A P value of less than .05 was considered statistically significant. significantly fewer morphine boluses in the K group than those in the P group (Table 2). The VAS score was not significantly different between 3. Results groups K and P at T0 and T30 (Table 2). Evolution of 3.1. Patient characteristics the VAS score variation (DVAS [Tx], defined as VAS [T0] À AS [Tx]) was not significantly different between Between January 01, 2004, and June 30, 2005, seventy- the groups (Fig. 2). At T30, 20 patients (61%) of the three patients were enrolled in the study (Fig. 1). Seven K group had a VAS score not exceeding 30/100 vs patients (5 in the K group and 2 in the P group) were 13 patients (41%) of the P group, representing a nonsignif- withdrawn from the analysis because of incomplete data or icant difference ( P = .2). disrespect of study protocol. One patient was excluded because of an anaphylactoid reaction after antibiotic 3.3. Secondary outcome injection in the P group. Thus, data from 65 patients were Satisfaction of patients was not significantly different completed and analyzed, 33 in the K group and 32 in between the 2 groups. There were no differences between the P group. groups with regard to blood pressure, heart rate, respiratory Baseline characteristics were similar between the rate, or oxygen saturation at T0 and T30 (Table 3). The 2 groups (Table 1). incidence of neuropsychological adverse effects was signif- 3.2. Primary outcome icantly greater in the K group, with hallucinations (n = 4), dizziness (n = 6), diplopia (n = 2), and dysphoria (n = 6), At T30, morphine consumption was significantly lower whereas there was only 1 case of dysphoria in the P group. in the K group than that in the P group, corresponding to Some patients had 2 or more symptoms. Table 2 Comparison of morphine requirements, number of morphine boluses, VAS, and patient satisfaction between groups K and P K group P group P (n = 33) (n = 32) Morphine requirements (mg d kgÀ1, mean [95% CI]) T0 0.099 (0.097-0.102) 0.096 (0.0917-0.100) .2 T30 0.149 (0.132-0.165) 0.202 (0.181-0.223) b.001 No. of morphine boluses (median [interquartile range]) 1.0 (0.0-2.0) 2.3 (1.8-3.8) b.0001 VAS (mean [95% CI]) T0 80.4 (75.6-85.2) 80.7 (75.8-85.6) NS T30 34.1 (25.6-42.6) 39.5 (32.4-46.6) NS Patient satisfaction T30 (n [%]), excellent or good 18 (56) 22 (69) 0.3 NS indicates not significant; CI, confidence interval.
  • 5. Management of severe acute pain in emergency settings 389 Table 3 Comparison between groups K and P for clinical results were similar whatever their designs, namely, that parameters and adverse effects small-dose ketamine reduced morphine requirements and pain intensity [5,6,12]. Weinbroum [12] evaluated the Parameters K group P group P effects of postoperative coadministration of small doses of (n = 33) (n = 32) ketamine (0.250 mg d kgÀ1) and morphine on pain intensity Heart rate in surgical patients who complained of pain of at least 6/10 (beats/min, mean F SD) on a VAS despite more than 0.1 mg d kgÀ1 of IV morphine T0 87 F 15 81 F 18 .3 T30 82 F 14 78 F 15 .5 administration within 30 minutes. This study demonstrated Systolic blood pressure that a single dose of 0.250 mg d kgÀ1 of ketamine plus (mm Hg, mean F SD) 0.015 mg d kgÀ1 of morphine provided analgesia in 68% of T0 139 F 17 139 F 21 .9 patients compared with only 3.5% of patients who received T30 136 F 20 133 F 20 .5 0.030 mg d kgÀ1 of morphine with saline. Pain intensity Respiratory rate was still lower in the former group 2 hours after. (breaths/min, mean F SD) The idea of combining opiates and NMDA receptor T0 20 19 .6 antagonists is based on numerous experimental data T30 18 17 .5 suggesting different and complementary mechanisms of Pulse oximetry action for these 2 classes of analgesic agents [21,22]. (%, mean F SD) Morphine acts at both the spinal and supraspinal levels. The T0 99 F 2 99 F 1 .8 T30 99 F 1 98 F 2 .2 spinal action is well documented [23]. It is generally No. of patients with 7 (21) 2 (6) .2 accepted that opioids reduce the spinal transmission of Ramsay score z3 (n [%]) nociceptive signals predominantly at presynaptic sites Adverse effects (n [%]) (ie, reduction in transmitter release from afferent C fibers), Nausea and vomiting 8 (6) 4 (6) NS although a postsynaptic inhibition of spinal dorsal horn Neuropsychological 12 (36) 1 (3) .002 nociceptive neurons has also been demonstrated. In contrast, Itching 1 (3) 1 (3) NS NMDA receptor antagonists preferentially act postsynapti- Bradypnea 0 1 (3) NS cally by reducing the hyperexcitability (ie, central sensiti- zation) of spinal nociceptive neurons [21,24]. Thus, the reduction in nociceptive inputs associated with the reduction The level of sedation, nausea, vomiting, and itching did in postsynaptic neuronal hyperexcitability may explain the not differ between the 2 groups (Table 3). synergistic interaction of the 2 drugs [8]. Bossard et al [8] analyzed the effects of morphine, ketamine, and their combination on electrophysiological recordings of the 4. Discussion nociceptive flexion RIII reflex in 12 healthy volunteers. The stimulus response curve of the nociceptive RIII reflex In trauma patients with severe acute pain, we found that was significantly reduced after injection of a combination of the association of low-dose ketamine with morphine ketamine and morphine, and they concluded there was a reduced morphine requirements by approximately 26% synergistic interaction between the 2 drugs. However, we within 30 minutes. However, pain intensity measured on a found that there were significantly more adverse effects VAS at T30 was not different between the 2 groups. To our with ketamine. Analgesic doses of 0.150 to 0.500 mg d kgÀ1 knowledge, this is the first study that associated morphine of ketamine have been reported to produce dose- or plasma with low-dose ketamine for trauma patients with severe concentration–dependent cognitive, perceptual, and mood acute pain in emergency settings. disturbances, as well as psychotomimetic adverse effects Gurnani et al [20] studied low-dose ketamine in patients [25,26]. This could be a limitation to the use of ketamine in with acute pain after musculoskeletal trauma in emergency such settings, especially if the benefit regarding pain is poor. settings. However, in this study, patients initially received In our study, all adverse effects were weak and brief because 0.250 mg d kgÀ1 of IV ketamine, followed by a constant-rate no one needed treatment and patients’ satisfaction was not infusion of subcutaneous ketamine (0.100 mg d kgÀ1 d hÀ1) different between the 2 groups. In a review article about without morphine, whereas the control group received IV perioperative ketamine for acute postoperative pain, Bell injections of morphine (0.1 mg d kgÀ1 every 4 hours). They et al [27] found that the occurrence of adverse effects was found that the onset of analgesia was slower in the morphine similar in ketamine- and placebo-treated groups. Especially, group and that analgesia was more intense in the ketamine 21 of 37 trials specifically stated that there were no group [20]. Furthermore, the rate of patients requiring psychotomimetic adverse effects. However, in an experi- supplementary analgesia was lower in the ketamine group. mental study on 12 healthy volunteers, Bossard et al found However, these patients did not have severe pain. many psychosensory effects (8-10/12 volunteers) after the Most studies about small doses of ketamine in painful administration of ketamine, morphine, or their combination, patients were performed in postoperative settings. Their but the incidence was similar between groups.
  • 6. 390 M. Galinski et al. The aim of a coanalgesic association is to avoid opioid [9] Sethna NF, Liu M, Gracely R, et al. Analgesic and cognitive effects of overdose and adverse effects with equal or better analgesia. intravenous ketamine-alfentanil combinations versus either drug alone after intradermal capsaicin in normal subjects. Anesth Analg 1998; Weinbroum [12] obtained significantly fewer adverse effects 86:1250 - 6. such as nausea and vomiting with ketamine and morphine [10] Kissin I, Bright CA, Bradley Jr EL. The effect of ketamine on opioid- compared with morphine and placebo (12% and 38%, induced acute tolerance: can it explain reduction of opioid consump- respectively). We did not reach this conclusion because it tion with ketamine-opioid analgesic combinations? Anesth Analg was not the first end point of our study and the patient 2000;91:1483 - 8. [11] Angst MS, Koppert W, Pahl I, et al. Short-term infusion of the A- number was too small for that. opioid agonist remifentanil in humans causes hyperalgesia during The observation period also has to be longer than withdrawal. Pain 2003;106:49 - 57. 30 minutes because there is probably a ketamine activity [12] Weinbroum AA. A single small dose of postoperative ketamine that lasts longer than its simple pharmacokinetic action provides rapid and sustained improvement in morphine analgesia particularly on hyperalgesia. This point was demonstrated in in the presence of morphine-resistant pain. Anesth Analg 2003; postoperative studies [12]. Furthermore, the ketamine we 96:789 - 95. [13] Bijur PE, Kenny MK, Gallagher EJ. Intravenous morphine at used was a racemic mixture consisting of equal shares of 0.1 mg/kg is not effective for controlling severe acute pain in the 2 optical enantiomers. Pharmacologic investigations show majority of patients. Ann Emerg Med 2005;46:362 - 5. differences between those enantiomers, with a clinical [14] Mao J, Price DD, Mayer DJ. Mechanisms of hyperalgesia and superiority of S(+)-ketamine particularly with fewer adverse morphine tolerance: a current view of their possible interaction. Pain effects [28]. Unfortunately, this enantiomer is not yet 1995;62:259 - 74. [15] Adnet F, Jouriles NJ, Le Toumelin P, Hennequin B, Taillandier C, available in our country. Rayeh F, et al. Survey of out-of-hospital emergency intubations in the The observation period of our trial is a limitation because French prehospital medical system: a multicenter study. Ann Emerg 30 minutes was probably too short. Indeed, Weinbroum [12] Med 1998;32:454 - 60. demonstrated that the analgesic effects of the ketamine [16] Ramsay MA, Savege TM, Simpson BR, Gooodwin R. Controlled group were clearly evident throughout the 120-minute sedation with alphaxalone-alphadolone. Br J Med 1974;2:256 - 9. [17] Ricard-Hibon A, Chollet C, Saada S, et al. A quality control program observation period. for acute pain management in out-of-hospital critical care medicine. Ann Emerg Med 1999;34:738 - 44. 5. Conclusion [18] Todd KH, Funk KG, Funk JP, Bonacci R. Clinical significance of reported changes in pain severity. Ann Emerg Med 1996;27: This study showed that low-dose ketamine in trauma 485 - 9. [19] Casagrande JT, Pike MC, Smith PG. An improved approximate patients with severe acute pain reduced morphine require- formula for calculating sample sizes for comparing two binomial ments. We could not demonstrate any reduction in pain distributions. Biometrics 1978;34:483 - 6. intensity with ketamine in this study. [20] Gurnani A, Sharma PK, Rautela RS, Bhattacharya A. Analgesia for acute musculoskeletal trauma: low-dose subcutaneous infusion of ketamine. Anaesth Intensive Care 1996;24:32 - 6. References [21] Dickenson AH. NMDA receptor antagonists: interactions with opioids. Acta Anaesthesiol Scand 1997;41:112 - 5. [1] Belpomme V, Ricard-Hibon A, Cholet C, et al. Delai d’obtention de ´ [22] Wiesenfeld-Hallin Z. Combined opioid-NMDA antagonist therapies. l’analgesie en prehospitalier. Ann Fr Anesth Reanim 2002;21:324S ´ ´ What advantages do they offer the control of pain syndromes? Drugs [abstract]. 1998;56:1 - 5. [2] Paqueron X, Lumbroso A, Mergoni P, et al. Is morphine-induced [23] Yaksh TL. Pharmacology and mechanisms of opioid analgesic sedation synonymous with analgesia during intravenous morphine activity. Acta Anaesthesiol Scand 1997;41:94 - 111. titration? Br J Anaesth 2002;89:697 - 701. [24] Chapman V, Dickenson AH. The combination of NMDA antagonism [3] Kehlet H, Dahl JB. The value of bmultimodalQ or bbalanced analgesiaQ and morphine produces profound antinociception in the rat dorsal in postoperative pain treatment. Anesth-Analg 1993;77:1048 - 56. horn. Brain Res 1992;573:321 - 3. [4] Richebe P, Rivat C, Rivalan B, Maurette P, Simonnet G. Low doses ´ [25] Oye I, Paulsen O, Maurset A. Effects of ketamine on sensory ketamine: antihyperalgesic drug, non-analgesic. Ann Fr Anesth perception: evidence for a role of N-methyl-d-aspartate receptors. Reanim 2005;24:11349 - 59. J Pharmacol Exp Ther 1992;260:1209 - 13. [5] Kapfer B, Alfonsi P, Guignard B, Sessler DI, Chauvin M. Nefopam [26] Krystal JH, Karper LP, Seibyl JP, et al. Subanesthetic effects of and ketamine comparably enhance postoperative analgesia. Anesth noncompetitive NMDA antagonist, ketamine, in humans: psychomi- Analg 2005;100:169 - 74. metic, perceptual, cognitive and neuroendocrine responses. Arch Gen [6] Suzuki M, Kentaro T, Lansing PS, Tolan MM, Fuhrman TM, Ignacio Psychiatry 1994;14:144 - 53. CI, et al. Small-dose ketamine enhances morphine-induced analgesia [27] Bell RF, Dahl JB, Moore RA, Kalso E. Perioperative ketamine after outpatient surgery. Anesth Analg 1999;89:98 - 103. for acute post-operative pain: a quantitative and qualitative systema- [7] Alvarez P, Saavedra G, Hernandez A, et al. Synergistic antinociceptive tic review (Cochrane review). Acta Anaesthesiol Scand 2005;49: effects of ketamine and morphine in the orofacial capsaicin test in the 1405 - 28. rat. Anesthesiology 2003;99:969 - 75. [28] Pfenninger EG, Durieux ME, Himmelseher S. Cognitive impairment [8] Bossard AE, Guirimand F, Fletcher D, et al. Interaction of a after small-dose ketamine isomer in comparison to equianalgesic combination of morphine and ketamine on the nociceptive flexion racemic ketamine in human volunteers. Anesthesiology 2002; reflex in human volunteers. Pain 2002;98:47 - 57. 96:357 - 66.