Journal articles on the topic 'Intravenous Ketamine'

To see the other types of publications on this topic, follow the link: Intravenous Ketamine.

Create a spot-on reference in APA, MLA, Chicago, Harvard, and other styles

Select a source type:

Consult the top 50 journal articles for your research on the topic 'Intravenous Ketamine.'

Next to every source in the list of references, there is an 'Add to bibliography' button. Press on it, and we will generate automatically the bibliographic reference to the chosen work in the citation style you need: APA, MLA, Harvard, Chicago, Vancouver, etc.

You can also download the full text of the academic publication as pdf and read online its abstract whenever available in the metadata.

Browse journal articles on a wide variety of disciplines and organise your bibliography correctly.

1

Zhang, Shi-Xia, Xin Li, Qing-Ming Ren, Dong-Liang Niu, Li Gao, and Hong-Bin Wang. "Changes of lidocaine concentration and physiological indices in dogs during anaesthesia with lidocaine and isoflurane combined with ketamine or fentanyl." Acta Veterinaria Brno 85, no. 1 (2016): 91–97. http://dx.doi.org/10.2754/avb201685010091.

Full text
Abstract:
Fentanyl and ketamine are often used as adjuvants in intravenous anaesthesia to prolong analgesia. The aim of this study was to compare changes of the basic physiological variables of intravenous lidocaine administration in combination with ketamine or fentanyl, and to evaluate the impact of addition of fentanyl or ketamine to lidocaine on serum lidocaine concentrations in dogs after intravenous administration. During general anaesthesia, dogs of group L received 2% lidocaine intravenously, dogs of group LF received 2% lidocaine and fentanyl, and dogs of the group LK received 2% lidocaine and ketamine. The heart rate, systolic arterial pressure, diastolic arterial pressure, mean arterial pressure and rectal temperature decreased in all groups, and group LF showed the biggest effect on the basic physiological variables, with the lowest heart rate during the test, significantly decreased rectal temperature, and the most decreased values of arterial pressure. Blood for determination of serum lidocaine concentration was taken before anaesthesia and 5, 30, 60, 90, 120, 150 and 180 min after initial intravenous injection of drugs. Fentanyl and ketamine did not cause significant changes of serum lidocaine concentration in dogs and may be used as adjuvant in intravenous anaesthesia without a significant increase in lidocaine absorption.
APA, Harvard, Vancouver, ISO, and other styles
2

Opler, Lewis A., Mark G. A. Opler, and Amy F. T. Arnsten. "Ameliorating treatment-refractory depression with intranasal ketamine: potential NMDA receptor actions in the pain circuitry representing mental anguish." CNS Spectrums 21, no. 1 (January 26, 2015): 12–22. http://dx.doi.org/10.1017/s1092852914000686.

Full text
Abstract:
This article reviews the antidepressant actions of ketamine, an N-methyl-D-aspartame glutamate receptor (NMDAR) antagonist, and offers a potential neural mechanism for intranasal ketamine’s ultra-rapid actions based on the key role of NMDAR in the nonhuman primate prefrontal cortex (PFC). Although intravenous ketamine infusions can lift mood within hours, the current review describes how intranasal ketamine administration can have ultra-rapid antidepressant effects, beginning within minutes (5–40 minutes) and lasting hours, but with repeated treatments needed for sustained antidepressant actions. Research in rodents suggests that increased synaptogenesis in PFC may contribute to the prolonged benefit of ketamine administration, beginning hours after administration. However, these data cannot explain the relief that occurs within minutes of intranasal ketamine delivery. We hypothesize that the ultra-rapid effects of intranasal administration in humans may be due to ketamine blocking the NMDAR circuits that generate the emotional representations of pain (eg, Brodmann Areas 24 and 25, insular cortex), cortical areas that can be overactive in depression and which sit above the nasal epithelium. In contrast, NMDAR blockade in the dorsolateral PFC following systemic administration of ketamine may contribute to cognitive deficits. This novel view may help to explain how intravenous ketamine can treat the symptoms of depression yet worsen the symptoms of schizophrenia.
APA, Harvard, Vancouver, ISO, and other styles
3

Jiang, S., K. Hu, HG Fan, BS Yin, X. Li, JL Hou, and HB Wang. "Effects of ketamine/xylazine premedication on emulsified isoflurane general anaesthesia in swine undergoing embryo transplantation." Veterinární Medicína 59, No. 7 (September 16, 2014): 325–30. http://dx.doi.org/10.17221/7618-vetmed.

Full text
Abstract:
Cardiorespiratory effects were assessed during ketamine/xylazine premedication followed by emulsified isoflurane anaesthesia in swine undergoing experimental embryo transplantation. Ketamine (10 mg/kg) and xylazine (3.5 mg/kg) were premedicated intravenously, followed by continuous administration of intravenous emulsified isoflurane (2.8 ml/kg/h). Cardiorespiratory parameters, including heart rate, respiratory rate, mean arterial blood pressure, arterial oxygen saturation, and rectal temperature, were recorded in sows undergoing surgical embryo transplantation. Ketamine/xylazine premedication resulted in anaesthetic induction and lateral recumbency within 1 minute without any adverse effects. The physiological changes observed after drug administration remained within biologically acceptable limits. In conclusion, the combination of ketamine/xylazine provided anaesthetic induction, muscle relaxation, and analgesia sufficient for emulsified isoflurane intravenous anaesthesia. There were no adverse events in the experimental animals. This finding supports the use of emulsified isoflurane following ketamine/xylazine premedication in pigs.
APA, Harvard, Vancouver, ISO, and other styles
4

Kornhall, Daniel, and Erik Waage Nielsen. "Failure of Ketamine Anesthesia in a Patient with Lamotrigine Overdose." Case Reports in Critical Care 2014 (2014): 1–3. http://dx.doi.org/10.1155/2014/916360.

Full text
Abstract:
Introduction.It is important to know which clinical situations prevent ketamine from working.Case Report.We present the case of the psychiatric inpatient who was admitted to our emergency department after ingesting a toxic dose of lamotrigine, unknown at that time. On admission, she was clearly in distress, displaying extreme agitation and violent ataxic movements. We opted to achieve sedation using intravenous ketamine boluses. Unexpectedly, after being injected with a total of 250 mg ketamine, our patient displayed no signs of dissociative anaesthesia.Discussion.There was no apparent reason for why ketamine failed, but an interaction between lamotrigine and ketamine was suspected. A literature search was performed. Very few articles describe interactions between lamotrigine and ketamine. Experimental studies, however, demonstrate how lamotrigine attenuates the neuropsychiatric effects of ketamine. Ketamine is classically described as an NMDA antagonist. Ketamine’s dissociative effects, however, are thought to be mediated by increased glutamate release via a pathway not dependent on NMDA receptors. Lamotrigine, on the other hand, is known to reduce cortical glutamate release.Conclusion.Lamotrigine reduces the glutamate release needed to mediate ketamine’s dissociative anaesthesia. This is important knowledge for anaesthesiologists in the emergency room where ketamine is often administered to unstable patients.
APA, Harvard, Vancouver, ISO, and other styles
5

Stahl, Stephen M. "Mechanism of action of dextromethorphan/quinidine: comparison with ketamine." CNS Spectrums 18, no. 5 (September 20, 2013): 225–27. http://dx.doi.org/10.1017/s109285291300062x.

Full text
Abstract:
ISSUE:Reports of rapid-onset but short-duration antidepressant effects in patients with treatment-resistant mood disorders after intravenous administration of ketamine have prompted efforts to find an agent with ketamine's properties that can be administered orally in repeated doses in order to sustain that action. One candidate for this is dextromethorphan, and here the pharmacologic mechanism of action is compared and contrasted with that of ketamine.
APA, Harvard, Vancouver, ISO, and other styles
6

Tindale, Rabina. "Intravenous ketamine in adults." Emergency Nurse 16, no. 5 (September 13, 2008): 4. http://dx.doi.org/10.7748/en.16.5.4.s9.

Full text
APA, Harvard, Vancouver, ISO, and other styles
7

Ball, Christine, and Rod Westhorpe. "Intravenous Induction Agents: Ketamine." Anaesthesia and Intensive Care 30, no. 2 (April 2002): 115. http://dx.doi.org/10.1177/0310057x0203000201.

Full text
APA, Harvard, Vancouver, ISO, and other styles
8

Park, J.-W., Y.-H. Jung, C.-W. Baek, H. Kang, and S.-M. Cha. "Effects of Low Dose Ketamine on Tourniquet-induced Haemodynamic Responses during General Anaesthesia." Journal of International Medical Research 35, no. 5 (September 2007): 600–608. http://dx.doi.org/10.1177/147323000703500504.

Full text
Abstract:
This study investigated the effect of a pre-operative low dose of intravenous ketamine on tourniquet-induced haemodynamic changes. Ten minutes after induction of general anaesthesia, 0.1 mg/kg ketamine in 10 ml of saline (ketamine group, n = 14) or 10 ml of normal saline (control group, n = 14) were administered intravenously. Systolic and diastolic blood pressures, and heart rate relative to tourniquet inflation and deflation were recorded and compared within and between groups. Systolic and diastolic blood pressures in the control group significantly increased relative to baseline during the observation period following tourniquet inflation, but generally did not significantly increase in the ketamine group. The control group had a greater percentage of patients with a 30% rise in blood pressure at 60 min after tourniquet inflation compared with the ketamine group (28.6% vs 7.1%), but this was not statistically significant. We conclude that a pre-operative low dose (0.1 mg/kg) of intravenous ketamine can prevent a systemic arterial pressure increase for at least 60 min after tourniquet inflation under general anaesthesia.
APA, Harvard, Vancouver, ISO, and other styles
9

Mwase, Richard, Tonny Stone Luggya, John Mark Kasumba, Humphrey Wanzira, Andrew Kintu, Joesph V. B. Tindimwebwa, and Daniel Obua. "Analgesic Effects of Preincision Ketamine on Postspinal Caesarean Delivery in Uganda’s Tertiary Hospital: A Randomized Clinical Trial." Anesthesiology Research and Practice 2017 (2017): 1–6. http://dx.doi.org/10.1155/2017/5627062.

Full text
Abstract:
Background. Good postoperative analgesic management improves maternal satisfaction and care of the neonate. Postoperative pain management is a challenge in Mulago Hospital, yet ketamine is accessible and has proven benefit. We determined ketamine’s postoperative analgesic effects.Materials and Methods. We did an RCT among consenting parturients that were randomized to receive either intravenous ketamine (0.25 mg/kg) or placebo after spinal anesthetic. Pain was assessed every 30 mins up to 24 hours postoperatively using the numerical rating scale. The first complaint of pain requiring treatment was noted as “time to first breakthrough pain.”Results. We screened 100 patients and recruited 88 that were randomized into two arms of 44 patients that received either ketamine or placebo. Ketamine group had 30-minute longer time to first breakthrough pain and lower 24-hour pain scores. Postoperative diclofenac consumption was lesser in the ketamine group compared to placebo and Kaplan-Meier graphs showed a higher probability of experiencing breakthrough pain earlier in the placebo group.Conclusion. Preincision intravenous ketamine (0.25 mg/kg) offered 30-minute prolongation to postoperative analgesia requirement with reduced 24-hour pain scores. We recommend larger studies to explore this benefit. This trial is registered with Pan African Clinical Trial Registry numberPACTR201404000807178.
APA, Harvard, Vancouver, ISO, and other styles
10

AHMED, FAROOQ. "(TIVA) INTRAVENOUS ANAESTHESIA." Professional Medical Journal 13, no. 03 (June 25, 2006): 341–43. http://dx.doi.org/10.29309/tpmj/2006.13.03.4978.

Full text
Abstract:
To compare the combination of propofol ketamine , propofolfentanyl and propofol vs midazolam for TIVA in terms of haemodynamic changes, analgesia and recoverycharacteristics. Design A randomized clinical control trial. Setting CMH Rawalpindi. Period. September 2002 toAugust 2003.Material and Methods. The present study consisted of 75 patients of both sexes between the age groupof 18-55 years belonging to ASA grade I and II, who were scheduled for various short surgical procedures. Results.Combination of propofol and ketamine provides better haemodynamic stability throughout the procedure, whencompared to propofol fentanyl. Induction time was significantly shorter in the propofol group. Mean recovery time wassignificantly faster with propofol (12.3 min) compared to midazolam (20.4 min).. Conclusion. Combination of propofoland ketamine gives better haemodynamic stability during induction and maintenance of total intravenous anaesthesia.Both propofol and midazolam are safe and effective for TIVA in children. Recovery was faster and more comfortablefollowing propofol
APA, Harvard, Vancouver, ISO, and other styles
11

Arthur Twohig, Patrick, and Vaughn Huckfeldt. "Using Oral and Intranasal Dosage Forms of Ketamine for Managing Treatment-Resistant Depression: A Review of the Literature." International Journal of Medical Students 4, no. 2 (July 27, 2016): 64–71. http://dx.doi.org/10.5195/ijms.2016.153.

Full text
Abstract:
A lack of effective treatment for patients with treatment-resistant depression (TRD) has led to the evaluation of ketamine, an N-methyl- D-aspartate receptor antagonist. Despite the demonstrated short-term benefits of using intravenous (IV) ketamine, side effects and the difficulty in administering ketamine outside the health-care setting has raised interest in alternative dosage forms. Research articles evaluating oral or intranasal (IN) ketamine were retrieved from the PubMed database. Patients who received oral or IN ketamine experienced a similar reduction in depressive symptoms within 24 hours of treatment and fewer side effects compared to patients who received IV ketamine. Novel administration forms of ketamine provide an opportunity for patients with TRD to achieve remission with fewer adverse side effects. Future studies should continue to evaluate these administration strategies in the hope of promoting ketamine’s use outside health-care settings and for longer time periods.
APA, Harvard, Vancouver, ISO, and other styles
12

Gómez-Revuelta, Marcos, María Fernández-Rodríguez, Laura Boada-Antón, and Javier Vázquez-Bourgon. "Apnea during slow sub-anaesthetic infusion of intravenous ketamine for treatment-resistant depression." Therapeutic Advances in Psychopharmacology 10 (January 2020): 204512532098149. http://dx.doi.org/10.1177/2045125320981498.

Full text
Abstract:
Ketamine’s pharmacological profile makes it an interesting and useful drug to challenge treatment-resistant-depression (TRD). Emerging adverse events associated with single-slow-sub-anaesthetic doses for the treatment of depression are common, although generally transient and self-limited. Nevertheless, data on the safety of this practice are scarce. Thus, it seems timely before ketamine is used for clinical treatment of depression to recommend careful monitoring and reporting of all potential adverse events related to ketamine administration. Here, we describe a case of apnea during slow sub-anaesthetic infusion of intravenous ketamine for the treatment of resistant depression. As far as we are concerned, this is an uncommon, previously unreported, and potentially severe adverse event that clinicians should be aware of, and specific management measures should be implemented.
APA, Harvard, Vancouver, ISO, and other styles
13

Cusin, Cristina, Dawn Flosnik Ionescu, Kara Jean Pavone, Oluwaseun Akeju, Paolo Cassano, Norman Taylor, Matthias Eikermann, et al. "Ketamine augmentation for outpatients with treatment-resistant depression: Preliminary evidence for two-step intravenous dose escalation." Australian & New Zealand Journal of Psychiatry 51, no. 1 (February 18, 2016): 55–64. http://dx.doi.org/10.1177/0004867416631828.

Full text
Abstract:
Objective: Preliminary evidence supports the safety and efficacy of subanesthetic ketamine as an experimental antidepressant, although its effects are often not sustained beyond one week. Studies are lacking that have examined the sustained effects of escalating ketamine doses as augmentation in outpatients with treatment-resistant depression. Therefore, the aims of this study were twofold: (1) to assess the safety and antidepressant efficacy of two-step, repeated-dose ketamine augmentation and (2) to assess the duration of ketamine’s antidepressant efficacy as augmentation to ongoing antidepressant pharmacotherapy for 3 months after the final infusion. Methods: Fourteen patients with treatment-resistant depression were eligible to receive augmentation with six open-label intravenous ketamine infusions over 3 weeks. For the first three infusions, ketamine was administered at a dose of 0.5 mg/kg over 45 minutes; the dose was increased to 0.75 mg/kg over 45 minutes for the subsequent three infusions. The primary outcome measure was response (as measured on Hamilton Depression Rating Scale–28 items). Results: After the completion of three ketamine infusions, 7.1% (1/14) responded; after all six ketamine infusions, 41.7% (5/12) completers responded and 16.7% (2/12) remitted. Intent-to-treat response and remission rates at the end of the final infusion were 35.7% (5/14) and 14.3% (2/14), respectively. However, all but one responder relapsed within 2 weeks after the final infusion. Conclusion: Repeated, escalating doses of intravenous ketamine augmentation were preliminarily found to be feasible, efficacious and well tolerated. Interaction with concomitant medications and elevated level of treatment resistance are possible factors for non-response.
APA, Harvard, Vancouver, ISO, and other styles
14

Santos, Gustavo José Von Glehn dos, Eduardo Hatschbach, Ewaldo de Mattos Júnior, and Flávio Massone. "Parametric evaluation of methotrimeprazine-midazolam-ketamine and methotrimeprazine-midazolam-ketamine-xylazine combination in dogs." Acta Cirurgica Brasileira 21, no. 5 (October 2006): 304–9. http://dx.doi.org/10.1590/s0102-86502006000500006.

Full text
Abstract:
PURPOSE: To evaluate the parameters of dogs anesthetized by different dissociative drugs protocols through continuous intravenous infusion. METHODS: Thirty healthy dogs of both sexes were assigned randomly to three groups (G1, G2, and G3). G1 was administered with methotrimeprazine as a pre-anesthetic medication, intravenously midazolam-ketamine as bolus for induction and midazolam-ketamine by continuous intravenous infusion for a 60 minute-period of maintenance. G2: the same as for G1. plus an increase in the midazolam dose during maintenance. G3: the same treatment as for G2, plus the addition of xylazine during maintenance. Immediately after induction the anesthetic maintenance started, and measures were taken 15 minutes after pre-medication, at 10 minutes intervals, during maintenance (M0 to M7). RESULTS: Bradycardia, atrioventricular blockage, bradypnea and hypoxemia were shown in G3. G1 and G2 showed a slight hypotension only. CONCLUSION: There were some advantages by using the continuous intravenous via: no parameters oscillation and reduction in the anesthetic recovery period. The increase in midazolam dose brought about little parametric variations which were greater when xylazine was used, with a consequent hypoxemia, bradyarrhytmia, and decrease in respiratory frequency and minute volume.
APA, Harvard, Vancouver, ISO, and other styles
15

Levanen, Jaakko, Marja-Leena Makela, and Harry Scheinin. "Dexmedetomidine Premedication Attenuates Ketamine-induced Cardiostimulatory Effects and Postanesthetic Delirium." Anesthesiology 82, no. 5 (May 1, 1995): 1117–25. http://dx.doi.org/10.1097/00000542-199505000-00005.

Full text
Abstract:
Background Dexmedetomidine is a new potent and highly selective alpha 2-adrenoceptor agonist with sedative-hypnotic and anesthetic sparing properties. Because of its sympathoinhibitory activity, it may prove useful in balancing the cardiostimulatory effects and attenuating the adverse central nervous system effects of ketamine. Methods A double-blind, randomized and comparative parallel-group study design was employed in 40 volunteers with ASA physical status 1 who were scheduled for elective superficial surgery under ketamine anesthesia. Dexmedetomidine (2.5 micrograms/kg, n = 20) or midazolam (0.07 mg/kg, n = 20) was administered intramuscularly 45 min before induction of anesthesia. Anesthesia was induced with 2 mg/kg ketamine intravenously, and muscle relaxation was achieved with vecuronium. After tracheal intubation, anesthesia was maintained with nitrous oxide/oxygen (2:1) and additional 1 mg/kg intravenous ketamine boluses according to clinical and cardiovascular criteria. Hypotension and bradycardia were treated by increasing the intravenous infusion rate of crystalloids and intravenous atropine, respectively. Sedative and anxiolytic properties, intra- and postoperative drug requirements, psychomotor and cognitive impairments, and cardiovascular effects were compared between the two groups. Results Dexmedetomidine and midazolam proved to have equal sedative and anxiolytic effects after intramuscular administration, but dexmedetomidine induced significantly less preoperative psychomotor impairment and less anterograde amnesia than did midazolam. Compared to midazolam, dexmedetomidine decreased the need for intraoperative ketamine and was more effective in reducing ketamine-induced adverse central nervous system effects. Dexmedetomidine also was superior to midazolam in attenuating the hemodynamic responses to intubation and the cardiostimulatory effects of ketamine in general, but it increased the incidence of intra- and postoperative bradycardia. Conclusions These results suggest that premedication with 2.5 micrograms/kg dexmedetomidine is effective in attenuating the cardiostimulatory and postanesthetic delirium effects of ketamine. However, because of its propensity to cause bradycardia, routine use of an anticholinergic drug should be considered.
APA, Harvard, Vancouver, ISO, and other styles
16

Salman, Jasim M., Jasim N. Al-Asadi, Husham H. Abdul-Ra’aoof, Jawad H. Ahmed, and Ali H. Reshak. "COMPARISON OF INTRAMUSCULAR VERSUS INTRAVENOUS KETAMINE FOR SEDATION IN CHILDREN UNDERGOING MAGNETIC RESONANCE IMAGING EXAMINATION." Wiadomości Lekarskie 76, no. 1 (January 2023): 198–204. http://dx.doi.org/10.36740/wlek202301127.

Full text
Abstract:
The aim: To compare efficacy of intramuscular (IM) versus intravenous (IV) ketamine for sedation in children undergoing brain MRI scanning in children. Materials and methods: Children who required elective brain MRI were selected for this study. They were randomly divided into two groups; group I received 1.5 mg/kg IV Ketamine and group II received 4 mg/kg IM ketamine. In each group supplementary 0.1 mg/kg midazolam intravenously before positioning on MRI table was given. Patients were monitored for pulse rate, SPO2, and respiratory wave. Results: Children who received IM ketamine had significantly shorter scan time and a greater success rate of sedation with first dose than the IV group. The proportions of scan interruption and scan repeat were significantly higher among the IV group than in the IM group. The scan time was longer among the IV group than in the IM group with significantly more scan interruption and repeat. Satisfaction with sedation as expressed by the technicians was significantly more in the IM group than in IV group (98.1% vs. 80.8%, P= 0.004). Conclusions: Intramuscular ketamine injection was predicted to have a better sedative success rate and takes less time to complete than intravenous admin¬istration. This makes IM ketamine more appealing in certain conditions.
APA, Harvard, Vancouver, ISO, and other styles
17

Filzek, U., U. Fischer, and J. Ferguson. "Intravenous anaesthesia in hoses: racemic ketamine versus S-(+)-ketamine." Pferdeheilkunde Equine Medicine 19, no. 5 (2003): 501–6. http://dx.doi.org/10.21836/pem20030507.

Full text
APA, Harvard, Vancouver, ISO, and other styles
18

Amiot, J. F., and J. H. Palacci. "Intravenous regional anaesthesia with ketamine." Anaesthesia 40, no. 9 (September 1985): 899–901. http://dx.doi.org/10.1111/j.1365-2044.1985.tb11056.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
19

Verheecke, G. "Intravenous regional anaesthesia with ketamine." Anaesthesia 41, no. 9 (September 1986): 969–70. http://dx.doi.org/10.1111/j.1365-2044.1986.tb12943.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
20

Palacci, J. H., and J. F. Awor. "Intravenous regional anaesthesia with ketamine." Anaesthesia 41, no. 9 (September 1986): 970. http://dx.doi.org/10.1111/j.1365-2044.1986.tb12944.x.

Full text
APA, Harvard, Vancouver, ISO, and other styles
21

Höfler, Julia, and Eugen Trinka. "Intravenous ketamine in status epilepticus." Epilepsia 59 (August 26, 2018): 198–206. http://dx.doi.org/10.1111/epi.14480.

Full text
APA, Harvard, Vancouver, ISO, and other styles
22

Durrani, Zia, Alon P. Winnie, Elemer K. Zsigmond, and Martin L. Burnett. "Ketamine for Intravenous Regional Anesthesia." Anesthesia & Analgesia 68, no. 3 (March 1989): 328???332. http://dx.doi.org/10.1213/00000539-198903000-00026.

Full text
APA, Harvard, Vancouver, ISO, and other styles
23

Madathil, Shamnad, Deena Thomas, Parijat Chandra, Ramesh Agarwal, M. Jeeva Sankar, Anu Thukral, and Ashok Deorari. "‘NOPAIN-ROP’ trial: Intravenous fentanyl and intravenous ketamine for pain relief during laser photocoagulation for retinopathy of prematurity (ROP) in preterm infants: A randomised trial." BMJ Open 11, no. 9 (September 2021): e046235. http://dx.doi.org/10.1136/bmjopen-2020-046235.

Full text
Abstract:
ObjectivesTo investigate if intravenous fentanyl or intravenous ketamine can provide adequate analgesia in preterm infants undergoing laser photocoagulation for retinopathy of prematurity (ROP).DesignOpen-label randomised trial.SettingTertiary care institution.ParticipantsPreterm infants who underwent laser photocoagulation for ROP.InterventionsInfants were randomised to receive fentanyl as intravenous bolus dose of 2 µg/kg, followed by an intravenous infusion of 1 µg/kg/hour increased to a maximum of 3 µg/kg/hour or intravenous ketamine as bolus dose of 0.5 mg/kg, followed by further intermittent intravenous bolus doses of 0.5 mg/kg to a maximum of 2 mg/kg in the initial phase and intravenous fentanyl (bolus of 2 µg/kg followed by infusion of 2 µg/kg/hour to a maximum of 5 µg/kg/hour) or intravenous ketamine (bolus dose of 1 mg/kg followed by intermittent bolus doses of 0.5 mg/kg to a maximum of 4 mg/kg) in the revised regimen phase.Main outcome measuresProportion of infants with adequate analgesia defined as the presence of both: (1) all the Premature Infant Pain Profile-Revised scores measured every 15 min less than seven and (2) proportion of the procedure time the infant spent crying less than 5%.Secondary outcomes included apnoea, cardiorespiratory or haemodynamic instability, feed intolerance and urinary retention requiring catheterisation during and within 24 hours following the procedure.ResultsA total of 97 infants were randomised (fentanyl=51, ketamine=46). The proportions of infants with adequate analgesia were 16.3% (95% CI 8.5% to 29%) with fentanyl and 4.5% (95% CI 1.3% to 15.1%) with ketamine. Ten infants (19.6%) in the fentanyl group and seven infants (15.2%) in the ketamine group had one or more side effects. In view of inadequate analgesia with both the regimens, the study steering committee recommended using a higher dose of intravenous fentanyl and intravenous ketamine. Consequently, we enrolled 27 infants (fentanyl=13, ketamine=14). With revised regimens, the proportions of infants with adequate analgesia were higher: 23.1% (95% CI 8.2% to 50.2%) with fentanyl and 7.1% (95% CI 1.3% to 31.5%) with ketamine. However, higher proportions of infants developed apnoea (n=4; 30.7%), need for supplemental oxygen (n=5, 38.4%) and change in cardiorespiratory scores (n=7; 53.8%) with fentanyl but none with ketamine.ConclusionsFentanyl-based and ketamine-based drug regimens provided adequate analgesia only in a minority of infants undergoing laser photocoagulation for ROP. More research is needed to find safe and effective regimens that can be employed in resource constrained settings.Trial registration numberCTRI/2018/03/012878.
APA, Harvard, Vancouver, ISO, and other styles
24

Cairns, Brian E., Shelly A. McErlane, Miguel C. Fragoso, and Peter J. Soja. "Tooth Pulp– and Facial Hair Mechanoreceptor–evoked Responses of Trigeminal Sensory Neurons Are Attenuated during Ketamine Anesthesia." Anesthesiology 91, no. 4 (October 1, 1999): 1025. http://dx.doi.org/10.1097/00000542-199910000-00023.

Full text
Abstract:
Background Evidence exists that ketamine, administered systemically using a dose required for inducing a state of anesthesia, may antagonize nociceptive but not innocuous input to lumbar dorsal horn neurons. However, it is unclear whether ketamine exerts this selective action on sensory inputs to trigeminal sensory neurons. The current study was undertaken to compare the responses evoked in trigeminal sensory neurons by electrical stimuli applied to the tooth pulp versus air-puff stimuli applied to facial hair mechanoreceptors (FHMs) during quiet wakefulness versus ketamine anesthesia. Methods Accordingly, responses of rostral trigeminal sensory nuclear complex (TSNC) and trigeminothalamic tract neurons evoked by tooth pulp (a source of small-diameter fiber input) and FHMs (a source of larger-diameter fiber input) were recorded extracellularly from chronically instrumented cats before, during, and after recovery from the anesthetic state induced by a single (2.2 mg/kg) intravenous injection of ketamine. Results Overall, tooth pulp-evoked responses of TSNC neurons were maximally suppressed by 50% within 5 min after the intravenous administration of ketamine. Ketamine also suppressed the FHM-evoked responses of TSNC and trigeminothalamic neurons by 45%. The time course of ketamine's suppressive action was equivalent for tooth pulp- and FHM-evoked responses. However, the recovery of tooth pulp-evoked TSNC neuronal responses at suprathreshold intensities was markedly prolonged compared with neuronal responses driven by threshold stimuli or FHM. Conclusions These electrophysiologic results in the chronically instrumented cat preparation indicate that a nonselective suppression of orofacial somatosensory information occurs during ketamine anesthesia. The prolonged recovery of suprathreshold responses of TSNC neurons mediated by small-diameter afferent fiber input may partly underlie the analgesic action of ketamine that is clinically relevant at subanesthetic doses.
APA, Harvard, Vancouver, ISO, and other styles
25

Schoevers, Robert A., Tharcila V. Chaves, Sonya M. Balukova, Marije aan Het Rot, and Rudie Kortekaas. "Oral ketamine for the treatment of pain and treatment-resistant depression." British Journal of Psychiatry 208, no. 2 (February 2016): 108–13. http://dx.doi.org/10.1192/bjp.bp.115.165498.

Full text
Abstract:
BackgroundRecent studies with intravenous (i.v.) application of ketamine show remarkable but short-term success in patients with MDD. Studies in patients with chronic pain have used different ketamine applications for longer time periods. This experience may be relevant for psychiatric indications.AimsTo review the literature about the dosing regimen, duration, effects and side-effects of oral, intravenous, intranasal and subcutaneous routes of administration of ketamine for treatment-resistant depression and pain.MethodSearches in PubMed with the terms ‘oral ketamine’, ‘depression’, ‘chronic pain’, ‘neuropathic pain’, ‘intravenous ketamine’, ‘intranasal ketamine’ and ‘subcutaneous ketamine’ yielded 88 articles. We reviewed all papers for information about dosing regimen, number of individuals who received ketamine, number of ketamine days per study, results and side-effects, as well as study quality.ResultsOverall, the methodological strength of studies investigating the antidepressant effects of ketamine was considered low, regardless of the route of administration. The doses for depression were in the lower range compared with studies that investigated analgesic use. Studies on pain suggested that oral ketamine may be acceptable for treatment-resistant depression in terms of tolerability and side-effects.ConclusionsOral ketamine, given for longer time periods in the described doses, appears to be well tolerated, but few studies have systematically examined the longer-term negative consequences. The short- and longer-term depression outcomes as well as side-effects need to be studied with rigorous randomised controlled trials.
APA, Harvard, Vancouver, ISO, and other styles
26

Smith, Samantha A., Casey T. Fitzpatrick, Courtney L. Olesky, and Ashley B. Litchfield. "Management of Ketamine Extravasation in a Pediatric Patient During Procedural Sedation." Journal of Pediatric Pharmacology and Therapeutics 27, no. 3 (March 1, 2022): 292–95. http://dx.doi.org/10.5863/1551-6776-27.3.292.

Full text
Abstract:
Ketamine is a commonly used intravenous and intramuscular medication for procedural sedation within pediatric emergency medicine. There is limited availability of data on the rate of absorption and use of subcutaneous ketamine administration. We describe the case of a 12-year-old male who was sedated after extravasation and subsequent absorption of ketamine 1 mg/kg from a peripheral intravenous line (PIV). Despite being an unintended route, absorption of subcutaneous ketamine resulted in satisfactory procedural sedation with no complications. Given limited data on subcutaneous ketamine pharmacokinetics, the aim of this case report is to present the observed absorption of subcutaneous ketamine due to extravasation of PIV during a pediatric procedural sedation.
APA, Harvard, Vancouver, ISO, and other styles
27

Qaisaruddin, Syed. "Herniotomy in Children Under Intravenous Ketamine and Local Anesthesia." New Indian Journal of Surgery 11, no. 2 (June 1, 2020): 179–81. http://dx.doi.org/10.21088/nijs.0976.4747.11220.16.

Full text
APA, Harvard, Vancouver, ISO, and other styles
28

Huda, Anwar ul, Raheel Minhas, and Mohammad Yasir. "Intravenous ketamine in gynaecological surgeries reduces pain score and opioid consumption." Journal of the Pakistan Medical Association 72, no. 12 (November 15, 2022): 2491–97. http://dx.doi.org/10.47391/jpma.5275.

Full text
Abstract:
Objective: To assess the effect of intravenous ketamine on postoperative pain control, opioid consumption, and the incidence of postoperative adverse events in gynaecological surgeries. Method: The systematic review and meta-analysis was conducted in July 2020 and the search was repeated in July 2021 to ensure accuracy. The review was registered with the International Prospective Register of Systematic Reviews (PROSPERO) in July 2020. The search, done on online databases Medline and Science Direct, comprised studies on patients who underwent general anaesthesia for gynaecological surgeries and received intravenous ketamine intraoperatively, and the findings included opioid consumption, postoperative pain control, and associated side-effects. Results: Of the 79 randomised controlled trials found, 9(11.4%) were subjected to meta-analysis. The use of intravenous ketamine reduced pain score at 2h (p=0.003) and 24h (p=0.002) postoperatively in gynaecological surgeries. In laparoscopic gynaecological surgeries, lower pain scores were reported at 1h (p=0.01) and 2h (p=0.002) hours postoperatively. Lower pain scores were reported at 24h postoperatively in open gynaecological surgeries (p=0.002). Intravenous ketamine increased the time to first-request analgesia postoperatively (p=0.03), and reduced postoperative 24h opioid consumption (p=0.002). Conclusion: The use of intravenous ketamine significantly reduced postoperative pain at 2h and 24h after gynaecological surgeries and at 1h and 2h after laparoscopic gynaecological surgeries. Key Words: Ketamine, Pain score, Gynaecological surgery, Opioid consumption.
APA, Harvard, Vancouver, ISO, and other styles
29

Le Nedelec, Martin, Paul Glue, Helen Winter, Chelsea Goulton, and Natalie J. Medlicott. "The effect of route of administration on the enantioselective pharmacokinetics of ketamine and norketamine in rats." Journal of Psychopharmacology 32, no. 10 (June 13, 2018): 1127–32. http://dx.doi.org/10.1177/0269881118780013.

Full text
Abstract:
Background: Ketamine has been shown to produce a rapid and potent antidepressant response in patients with treatment-resistant depression. Currently ketamine is most commonly administered as a 40-minute intravenous infusion, though it is unknown whether this is the optimal route of administration. Aims: To determine the plasma concentration time course of the R- and S-enantiomers of ketamine and norketamine following administration of ketamine by four different routes of administration. Methods: Plasma from conscious non-anaesthetised rats was collected following administration of ketamine by either subcutaneous (SC), intramuscular (IM), intravenous infusion (IVI) or intravenous bolus (IVB) routes of administration. Concentrations of the enantiomers of ketamine and norketamine were determined by LC/MS. Results: Administration by the SC, IM and IVI routes produced an overall similar drug exposure. In contrast, administration by the IVB route produced approximately 15-fold higher peak plasma concentrations for the enantiomers of ketamine and an approximately four-fold lower AUC for the enantiomers of norketamine. Conclusions: Route of administration can significantly influence ketamine and norketamine exposures. These differences may influence safety and tolerability, and potentially drug efficacy in humans. This knowledge adds to current research into the optimisation of the use of ketamine for the treatment of depression.
APA, Harvard, Vancouver, ISO, and other styles
30

Wadhwa, Anupama. "Intraoperative Intravenous Methadone and Ketamine Combination versus Intravenous Morphine and Ketamine Combination for Post-Operative Analgesia in Patients Undergoing Lower Extremity Fracture Surgery." Clinical Research and Clinical Trials 3, no. 4 (May 28, 2021): 01–06. http://dx.doi.org/10.31579/2693-4779/026.

Full text
Abstract:
Background: Pain management for lower extremity fracture surgeries can be challenging. The purpose of this study is to determine whether the use of ketamine and methadone are more effective than ketamine and morphine to reduce postoperative pain and morphine requirements in patients undergoing lower extremity fracture surgery. Materials and Methods: Seventy-five patients 18-65 years of age, ASA class I-III, were enrolled in this study, which scheduled for elective lower extremity orthopedic surgery involving fracture of femur or tibia were recruited for the study. Thirty-eight randomized to the Methadone group and 37 randomized into the Morphine group. Participants were randomized to either one of the two groups: methadone (2ug/kg fentanyl, 0.2 mg/kg ketamine and 0.2 mg/kg methadone IV) versus control (2 ug/kg fentanyl, 0.2mg/kg ketamine and 0.2 mg/kg morphine IV). The primary outcome was total morphine equivalent (MEQ) during the first 24 and 48 hours after surgery. Secondary outcomes included postoperative pain scores in PACU, at 24 and 48 hours, as well as postoperative nausea and vomiting (PONV). Results: There was no difference in intraoperative consumption of fentanyl between the Methadone group 360mcg and Morphine group 344mcg. In the first 24 hours postoperatively, the Methadone group consumed less MEQ compared with the Morphine group (36.1 mg vs 54.8 mg, p=0.0072), showed lower pain scores than the Morphine group (p=0.0146), and experienced more nausea and vomiting than the Morphine group. There were no differences in sedation in both groups. Conclusion: The intraoperative use of intravenous methadone significantly reduced post-operative opioid requirement in patients undergoing lower extremity fracture surgery. The results also demonstrated the methadone group had a higher rate of PONV.
APA, Harvard, Vancouver, ISO, and other styles
31

O’Brien, Lijffijt, Wells, Swann, and Mathew. "The Impact of Childhood Maltreatment on Intravenous Ketamine Outcomes for Adult Patients with Treatment-Resistant Depression." Pharmaceuticals 12, no. 3 (September 11, 2019): 133. http://dx.doi.org/10.3390/ph12030133.

Full text
Abstract:
Childhood maltreatment is associated with a poor treatment response to conventional antidepressants and increased risk for treatment-resistant depression (TRD). The N-methyl-D-aspartate receptor (NDMAR) antagonist ketamine has been shown to rapidly improve symptoms of depression in patients with TRD. It is unknown if childhood maltreatment could influence ketamine’s treatment response. We examined the relationship between childhood maltreatment using the Childhood Trauma Questionnaire (CTQ) and treatment response using the Quick Inventory of Depressive Symptoms–Self Report (QIDS-SR) in TRD patients receiving intravenous ketamine at a community outpatient clinic. We evaluated treatment response after a single infusion (n = 115) and a course of repeated infusions (n = 63). Repeated measures general linear models and Bayes factor (BF) showed significant decreases in QIDS-SR after the first and second infusions, which plateaued after the third infusion. Clinically significant childhood sexual abuse, physical abuse, and cumulative clinically significant maltreatment on multiple domains (maltreatment load) were associated with better treatment response to a single and repeated infusions. After repeated infusions, higher load was also associated with a higher remission rate. In contrast to conventional antidepressants, ketamine could be more effective in TRD patients with more childhood trauma burden, perhaps due to ketamine’s proposed ability to block trauma-associated behavioral sensitization.
APA, Harvard, Vancouver, ISO, and other styles
32

Mishra, Priyanka. "A Case of Bradycardia and Hypersensitivity with Intravenous Ketamine Monotherapy." Clinical Medical Reviews and Reports 3, no. 2 (March 17, 2021): 01–03. http://dx.doi.org/10.31579/2690-8794/066.

Full text
Abstract:
Anaphylaxis during anesthesia is an unforeseeable and potentially life threatening syndrome that is dose independent. Ketamine is a widely used hypnotic for procedural sedation in the emergency department, in anesthesia and intensive care units (ICU). It is popularly employed for both children and adult patients. Though, dose dependent adverse effects of ketamine have been described, the hypersensitive reactions with the same are extremely rare. We are hereby presenting a case of an allergic reaction and isolated bradycardia with ketamine, given as intravenous monotherapy in a patient with no previous history of atopic disease. Our article aims at reminding all the medical professionals to never let a drug (however commonly used) throw away caution, be vigilant to timely diagnose an adverse drug reaction (even the most infrequent and rare) and be prepared to tackle the worst with the necessary supportive measures like hemodynamic support and advanced airway procedures.
APA, Harvard, Vancouver, ISO, and other styles
33

Radvansky, Brian M., Khushbu Shah, Anant Parikh, Anthony N. Sifonios, Vanny Le, and Jean D. Eloy. "Role of Ketamine in Acute Postoperative Pain Management: A Narrative Review." BioMed Research International 2015 (2015): 1–10. http://dx.doi.org/10.1155/2015/749837.

Full text
Abstract:
Objectives. The objective of this narrative review was to examine the usage of ketamine as a postoperative analgesic agent across a wide variety of surgeries.Design. A literature search was performed using the phrases “ketamine” and “postoperative pain.” The authors analyzed the studies that involved testing ketamine’s effectiveness at controlling postoperative pain. Effectiveness was assessed through various outcomes such as the amount of opiate consumption, visual analog scale (VAS) pain scores, and persistent postoperative pain at long-term follow-up.Results. While many different administration protocols were evaluated, delivering ketamine both as a pre- or perioperative bolus and postoperative infusion for up to 48 hours appeared to be the most effective. These effects are dose-dependent. However, a number of studies analyzed showed no benefit in using ketamine versus placebo for controlling postoperative pain. While ketamine is a safe and well-tolerated drug, it does have adverse effects, and there are concerns for possible neurotoxicity and effects on memory.Conclusions. In a number of limited situations, ketamine has shown some efficacy in controlling postoperative pain and decreasing opioid consumption. More randomized controlled trials are necessary to determine the surgical procedures and administrations (i.e., intravenous, epidural) that ketamine is best suited for.
APA, Harvard, Vancouver, ISO, and other styles
34

Suzuki, Manzo, Syuji Haraguti, Kikuzo Sugimoto, Takehiko Kikutani, Yoichi Shimada, and Atsuhiro Sakamoto. "Low-dose Intravenous Ketamine Potentiates Epidural Analgesia after Thoracotomy." Anesthesiology 105, no. 1 (July 1, 2006): 111–19. http://dx.doi.org/10.1097/00000542-200607000-00020.

Full text
Abstract:
Background Ketamine potentiates intravenous or epidural morphine analgesia. The authors hypothesized that very-low-dose ketamine infusion reduces acute and long-term postthoracotomy pain. Methods Forty-nine patients scheduled to undergo open thoracotomy were randomly assigned to receive one of two anesthesia regimens: continuous epidural infusion of ropivacaine and morphine, along with intravenous infusion of ketamine (0.05 mg . kg(-1) . h(-1) [approximately 3 mg/h], ketamine group, n = 24) or placebo (saline, control group, n = 25). Epidural analgesia was continued for 2 days after surgery, and infusion of ketamine or placebo was continued for 3 days. Pain was assessed at 6, 12, 24, and 48 h after surgery. Patients were asked about their pain, abnormal sensation on the wound, and inconvenience in daily life at 7 days and 1, 3, and 6 months after surgery. Results The visual analog scale scores for pain at rest and on coughing 24 and 48 h after thoracotomy were lower in the ketamine group than in the control group (pain at rest, 9 +/- 11 vs. 25 +/- 20 and 9 +/- 11 vs. 18 +/- 13; pain on coughing, 26 +/- 16 vs. 50 +/- 17 and 30 +/- 18 vs. 43 +/- 18, mean +/- SD; P = 0.002 and P = 0.01, P < 0.0001 and P = 0.02, respectively). The numerical rating scale scores for baseline pain 1 and 3 months after thoracotomy were significantly lower in the ketamine group (0.5 [0-4] vs. 2 [0-5] and 0 [0-5] vs. 1.5 [0-6], median [range], respectively; P = 0.02). Three months after surgery, a higher number of control patients were taking pain medication (2 vs. 9; P = 0.03). Conclusions Very-low-dose ketamine (0.05 mg . kg(-1) . h(-1)) potentiated morphine-ropivacaine analgesia and reduced postthoracotomy pain.
APA, Harvard, Vancouver, ISO, and other styles
35

Noppers, Ingeborg, Erik Olofsen, Marieke Niesters, Leon Aarts, René Mooren, Albert Dahan, Evan Kharasch, and Elise Sarton. "Effect of Rifampicin on S-ketamine and S-norketamine Plasma Concentrations in Healthy Volunteers after Intravenous S-ketamine Administration." Anesthesiology 114, no. 6 (June 1, 2011): 1435–45. http://dx.doi.org/10.1097/aln.0b013e318218a881.

Full text
Abstract:
Background Low-dose ketamine is used as analgesic for acute and chronic pain. It is metabolized in the liver to norketamine via cytochrome P450 (CYP) enzymes. There are few human data on the involvement of CYP enzymes on the elimination of norketamine and its possible contribution to analgesic effect. The aim of this study was to investigate the effect of CYP enzyme induction by rifampicin on the pharmacokinetics of S-ketamine and its major metabolite, S-norketamine, in healthy volunteers. Methods Twenty healthy male subjects received 20 mg/70 kg/h (n = 10) or 40 mg/70 kg/h (n = 10) intravenous S-ketamine for 2 h after either 5 days oral rifampicin (once daily 600 mg) or placebo treatment. During and 3 h after drug infusion, arterial plasma concentrations of S-ketamine and S-norketamine were obtained at regular intervals. The data were analyzed with a compartmental pharmacokinetic model consisting of three compartments for S-ketamine, three sequential metabolism compartments, and two S-norketamine compartments using the statistical package NONMEM® 7 (ICON Development Solutions, Ellicott City, MD). Results Rifampicin caused a 10% and 50% reduction in the area-under-the-curve of the plasma concentrations of S-ketamine and S-norketamine, respectively. The compartmental analysis indicated a 13% and 200% increase in S-ketamine and S-norketamine elimination from their respective central compartments by rifampicin. Conclusions : A novel observation is the large effect of rifampicin on S-norketamine concentrations and indicates that rifampicin induces the elimination of S-ketamine's metabolite, S-norketamine, probably via induction of the CYP3A4 and/or CYP2B6 enzymes.
APA, Harvard, Vancouver, ISO, and other styles
36

Hasan, Mahmoud, Christiane Modess, Tarek Roustom, Anne Dokter, Markus Grube, Andreas Link, Hélène Rey, Stefanie Adler, Konrad Meissner, and Werner Siegmund. "Chiral Pharmacokinetics and Metabolite Profile of Prolonged-release Ketamine Tablets in Healthy Human Subjects." Anesthesiology 135, no. 2 (May 21, 2021): 326–39. http://dx.doi.org/10.1097/aln.0000000000003829.

Full text
Abstract:
Background The anesthetic ketamine after intravenous dosing is nearly completely metabolized to R- and S-stereoisomers of the active norketamine (analgesic, psychoactive) and 2,6-hydroxynorketamine (potential analgesic, antidepressant) as well as the inactive dehydronorketamine. Oral administration favors the formation of 2,6-hydroxynorketamines via extensive presystemic metabolism. The authors hypothesized that plasma exposure to 2,6-hydroxynorketamines relative to the psychoactive ketamine is greater after prolonged-release ketamine tablets than it is after intravenous ketamine. Methods Pharmacokinetics of ketamine after intravenous infusion (5.0 mg) and single-dose administrations of 10, 20, 40, and 80 mg prolonged-released tablets were evaluated in 15 healthy white human subjects by means of a controlled, ascending-dose study. The stereoisomers of ketamine and metabolites were quantified in serum and urine by validated tandem mass-spectrometric assays and evaluated by noncompartmental pharmacokinetic analysis. Results After 40 mg prolonged-release tablets, the mean ± SD area under the concentrations–time curve ratios for 2,6-hydroxynorketamine/ketamine were 18 ± 11 (S-stereoisomers) and 30 ± 16 (R-stereoisomers) compared to 1.7 ± 0.8 and 3.1 ± 1.4 and after intravenous infusion (both P < 0.001). After 10 and 20 mg tablets, the R-ratios were even greater. The distribution volumes at steady state of S- and R-ketamine were 6.6 ± 2.2 and 5.6 ± 2.1 l/kg, terminal half-lives 5.2 ± 3.4 and 6.1 ± 3.1 h, and metabolic clearances 1,620 ± 380 and 1,530 ± 380 ml/min, respectively. Bioavailability of the 40 mg tablets was 15 ± 8 (S-isomer) and 19 ± 10% (R-isomer) and terminal half-life 11 ± 4 and 10 ± 4 h. About 7% of the dose was renally excreted as S-stereoisomers and 17% as R-stereoisomers. Conclusions Prolonged-release ketamine tablets generate a high systemic exposure to 2,6-hydroxynorketamines and might therefore be an efficient and safer pharmaceutical dosage form for treatment of patients with chronic neuropathic pain compared to intravenous infusion. Editor’s Perspective What We Already Know about This Topic What This Article Tells Us That Is New
APA, Harvard, Vancouver, ISO, and other styles
37

Flood, Pamela, and Matthew D. Krasowski. "Intravenous Anesthetics Differentially Modulate Ligand-gated Ion Channels." Anesthesiology 92, no. 5 (May 1, 2000): 1418–25. http://dx.doi.org/10.1097/00000542-200005000-00033.

Full text
Abstract:
Background Heteromeric neuronal nicotinic acetylcholine receptors (nAChRs) are potently inhibited by volatile anesthetics, but it is not known whether they are affected by intravenous anesthetics. Ketamine potentiates gamma-aminobutyric acid type A (GABAA) receptors at high concentrations, but it is unknown whether there is potentiation at clinically relevant concentrations. Information about the effects of intravenous anesthetics with different behavioral profiles on specific ligand-gated ion channels may lead to hypotheses as to which ion channel effect produces a specific anesthetic behavior. Methods A heteromeric nAChR composed of alpha4 and beta4 subunits was expressed heterologously in Xenopus laevis oocytes. Using the two-electrode voltage clamp technique, peak ACh-gated current was measured before and during application of ketamine, etomidate, or thiopental. The response to GABA of alpha1beta2gamma2s GABAA receptors expressed in human embryonic kidney cells and Xenopus oocytes was compared with and without coapplication of ketamine from 1 microm to 10 mm. Results Ketamine caused potent, concentration-dependent inhibition of the alpha4beta4 nAChR current with an IC50 of 0.24 microm. The inhibition by ketamine was use-dependent; the antagonist was more effective when the channel had been opened by agonist. Ketamine did not modulate the alpha1beta2gamma2s GABAA receptor response in the clinically relevant concentration range. Thiopental caused 27% inhibition of ACh response at its clinical EC50. Etomidate did not modulate the alpha4beta4 nAChR response in the clinically relevant concentration range, although there was inhibition at very high concentrations. Conclusions The alpha4beta4 nAChR, which is predominantly found in the central nervous system (CNS), is differentially affected by clinically relevant concentrations of intravenous anesthetics. Ketamine, commonly known to be an inhibitor at the N-methyl-D-aspartate receptor, is also a potent inhibitor at a central nAChR. It has little effect on a common CNS GABAA receptor in a clinically relevant concentration range. Interaction between ketamine and specific subtypes of nAChRs in the CNS may result in anesthetic behaviors such as inattention to surgical stimulus and in analgesia. Thiopental causes minor inhibition at the alpha4beta4 nAChR. Modulation of the alpha4beta4 nAChR by etomidate is unlikely to be important in anesthesia practice based on the insensitivity of this receptor to clinically used concentrations.
APA, Harvard, Vancouver, ISO, and other styles
38

Aida, Sumihisa, Tomohiro Yamakura, Hiroshi Baba, Kiichiro Taga, Satoru Fukuda, and Koki Shimoji. "Preemptive Analgesia by Intravenous Low-dose Ketamine and Epidural Morphine in Gastrectomy." Anesthesiology 92, no. 6 (June 1, 2000): 1624–30. http://dx.doi.org/10.1097/00000542-200006000-00020.

Full text
Abstract:
Background Morphine and ketamine may prevent central sensitization during surgery and result in preemptive analgesia. The reliability of preemptive analgesia, however, is controversial. Methods Gastrectomy patients were given preemptive analgesia consisting of epidural morphine, intravenous low-dose ketamine, and combinations of these in a randomized, double-blind manner. Postsurgical pain intensity was rated by a visual analog scale, a categoric pain evaluation, and cumulative morphine consumption. Results Preemptive analgesia by epidural morphine and by intravenous low-dose ketamine were significantly effective but not definitive. With epidural morphine, a significant reduction in visual analog scale scores at rest was observed at 24 and 48 h, and morphine consumption was significantly lower at 6 and 12 h, compared with control values. With intravenous ketamine, visual analog scale scores at rest and morphine consumption were significantly lower at 6, 12, 24, and 48 h than those in control subjects. The combination of epidural morphine and intravenous ketamine provided definitive preemptive analgesia: Visual analog scale scores at rest and morphine consumption were significantly the lowest at 6, 12, 24, and 48 h, and the visual analog scale score during movement and the categoric pain score also were significantly the lowest among the groups. Conclusion The results suggest that for definitive preemptive analgesia, blockade of opioid and N-methyl-d-aspartate receptors is necessary for upper abdominal surgery such as gastrectomy; singly, either treatment provided significant, but not definitive, postsurgical pain relief. Epidural morphine may affect the spinal cord segmentally, whereas intravenous ketamine may block brain stem sensitization via the vagus nerve during upper abdominal surgery.
APA, Harvard, Vancouver, ISO, and other styles
39

Djafar, Zulfikar, and A. Husni Tanra. "Pain Management after Bilateral Mastectomy Surgery with Continued Intravenous Ketamine: A Case Report." Journal of Anesthesiology and Clinical Research 4, no. 1 (November 16, 2022): 370–74. http://dx.doi.org/10.37275/jacr.v4i1.263.

Full text
Abstract:
Introduction: Ketamine is recommended as postoperative analgesia because ketamine is a classic anesthetic agent that is available in almost all hospitals, including hospitals with limited resources. This study aims to describe the continuous use of intravenous ketamine as postoperative pain management in a bilateral mastectomy. Case presentation: A woman, 35 years old, was admitted to the hospital with complaints of lumps in both breasts. On physical examination, the patient looked weak, with blood pressure 120/70 mmHg, pulse 105x/minute, respiratory rate 18x/minute, temperature 36.8ºC, and numeric rating scale (NRS) 7/10. In the thoracic region, mammary dextra, there is a lump the size of a fixed tennis ball, 15 cm in diameter, and the skin around the lump looks red with a dry wound. On palpation, there is tenderness and hardness; mamma sinistra has a lump the size of a melon, 20 cm in diameter, with an ulcer that is still wet. On palpation, there is tenderness and hardness, and fixation. The patient was diagnosed as mammary tumor dextra et sinistra, suspected malignancy, and a simple bilateral mastectomy was performed. The anesthetic technique used during the operation was general anesthesia with endotracheal intubation (GETA). The premedication given was dexamethasone 10 mg intravenously, midazolam 3 mg intravenously, and fentanyl 100 mcg intravenously. Postoperative pain management using ketamine 0.15 mg/kgBW was given a bolus, followed by 0.1 mg/kgBW/hour. Conclusion: The use of ketamine in postoperative bilateral mastectomy can reduce pain intensity and have minimal side effects.
APA, Harvard, Vancouver, ISO, and other styles
40

Kim, Dong Hee, and Sang Chul Lee. "Preemptive Effect of Preoperative Intravenous Ketamine." Korean Journal of Anesthesiology 37, no. 1 (1999): 100. http://dx.doi.org/10.4097/kjae.1999.37.1.100.

Full text
APA, Harvard, Vancouver, ISO, and other styles
41

Sear, John W. "Intravenous hypnotics: propofol, ketamine, and eltanolone." Current Opinion in Anaesthesiology 9, no. 4 (August 1996): 289–94. http://dx.doi.org/10.1097/00001503-199608000-00004.

Full text
APA, Harvard, Vancouver, ISO, and other styles
42

Whiteside, Jonathan. "Opioid sparing effects of intravenous ketamine." Acute Pain 2, no. 3 (September 1999): 152. http://dx.doi.org/10.1016/s1366-0071(99)80008-4.

Full text
APA, Harvard, Vancouver, ISO, and other styles
43

Pershad, Jay. "Intravenous Ketamine Bolus: Not So Fast!" Annals of Emergency Medicine 65, no. 6 (June 2015): 649–51. http://dx.doi.org/10.1016/j.annemergmed.2014.12.034.

Full text
APA, Harvard, Vancouver, ISO, and other styles
44

Kapural, Leo. "Opioid-Sparing Effect of Intravenous Outpatient Ketamine Infusions Appears Short-Lived in Chronic-Pain Patients with High Opioid Requirements." Pain Physician 4;13, no. 4;7 (July 14, 2010): 389–94. http://dx.doi.org/10.36076/ppj.2010/13/389.

Full text
Abstract:
Background and Objectives: Ketamine is opioid-sparing. It attenuates the onset of opioid tolerance, and suppresses opioid-induced hyperalgesia. This study evaluated whether or not repeated outpatient infusions of intravenous ketamine reduced the amount of pain and the amount of opioid requirements for patients suffering with chronic, noncancerous pain. Study Design: Retrospective study Setting: Outpatient pain clinic Methods: We reviewed the records of 18 patients taking high doses of opioids chronically and nonetheless reporting poorly controlled pain. A comparison control group of 18 similar patients with high opioid requirements who were not given ketamine were selected from our clinic population. Intervention: Intravenous ketamine infusions Measurement: VAS pain scores and opioid use Results: Morphometric and demographic characteristics, baseline opioid use, and pain scores were similar in the ketamine and comparison groups. Five patients given ketamine experienced no benefit and discontinued treatment after 1-2 infusions. One patient developed a supraventricular arrhythmia which immediately resolved upon cessation of the infusion. And another, despite pain relief, felt overly-anxious and opted out. Eleven patients thus completed 3-6 weekly ketamine infusions. At 6 months, 5 patients maintained less than 50% of their baseline opioid use, while the remaining patients returned to the baseline opioid use or increased their requirements. There was no significant difference in pain scores at 6 months in patients who received ketamine infusions and control group patients. Limitations: Retrospective nature of the study Conclusions: Outpatient intravenous ketamine infusions did not improve long-term pain scores in patients with high opioid requirements and only a few were able to substantially reduce opioid use. Considering infusion risks and cost of such outpatient treatment, ketamine infusions do not appear to be a feasible option for improving pain relief and decreasing opioid use in high-opioid requirement patients. Key words: Ketamine; anesthesia; chronic pain; opioids; continuous infusion; opioid requirements
APA, Harvard, Vancouver, ISO, and other styles
45

Maria I Dalamagka. "Combined Spinal-Epidural (CSE) technique plus ketamine in research laparotomies with vertical incision: Case report." GSC Advanced Research and Reviews 10, no. 3 (March 30, 2022): 176–78. http://dx.doi.org/10.30574/gscarr.2022.10.3.0073.

Full text
Abstract:
The combined spinal-epidural (CSE) technique, a comparatively new anesthetic choice, includes an initial subarachnoid injection followed by epidural catheter placement and subsequent administration of epidural medications. Clinical studies have demonstrated that the CSE technique provides excellent surgical conditions as quickly as with single-shot subarachnoid block conditions that are better than with epidural block alone. A 72-year-old man with pelvic tumor undergoing laparotomy surgery underwent combined regional anesthesia (Th12 - O1) plus small doses of Ketamine via intravenus infusion. The combined spinal-epidural technique along with intravenous Ketamine offers a better analgesic and hemodynamic result.
APA, Harvard, Vancouver, ISO, and other styles
46

Salvon, Jennifer H. "Intravenous Anesthetic Agents." Journal of Pharmacy Practice 7, no. 1 (February 1994): 13–21. http://dx.doi.org/10.1177/089719009400700104.

Full text
Abstract:
Intravenous (IV) anesthetics are used in the operating room setting for the induction and maintenance of general anesthesia. These agents are used in combination with many other therapeutic agents including inhalational anesthetics, anticholinergics, neuromuscular blockers, local anesthetics, and antihistamines. Currently available intravenous anesthetics include barbiturates (eg, thiopental and methohexital), benzodiazepines (eg, diazepam and midazolam), etomidate, ketamine, propofol, and opioids (eg, morphine, meperidine, fentanyl, alfentanil, and sufentanil). The barbiturates are the most frequently used agents for induction today. The benzodiazepines are used primarily for preoperative sedation intraoperatively; however, they may also be used for induction in certain clinical situations. Etomidate offers the advantage of minimal cardiovascular side effects, while ketamine is distinguished by stimulant cardiovascular effects. Propofol has a short recovery time and a low incidence of nausea and vomiting when compared with that of barbiturates and opioids. The opioids are used most often for the production of analgesia, although they may also be used as primary anesthetic agents in select patient populations. In this article, the pharmacokinetics of these agents will be reviewed as well as factors affecting their pharmacokinetic profile. These factors include age, hemodynamic changes, renal or hepatic dysfunction, and interaction with concomitant medications.
APA, Harvard, Vancouver, ISO, and other styles
47

Patanwala, Asad E., Jennifer R. Martin, and Brian L. Erstad. "Ketamine for Analgosedation in the Intensive Care Unit: A Systematic Review." Journal of Intensive Care Medicine 32, no. 6 (December 8, 2015): 387–95. http://dx.doi.org/10.1177/0885066615620592.

Full text
Abstract:
Objective: To evaluate the evidence for the use of intravenous ketamine for analgosedation in the intensive care unit. Methods: MEDLINE and EMBASE were queried from inception until July 2015. Search terms used included ketamine, intensive care, and critical care. The search retrieved 584 articles to be screened for inclusion. The intent was to include randomized controlled studies using sustained intravenous infusions (>24 hours) of ketamine in the critically ill patients. Results: One trial evaluated opioid consumption as an outcome in postoperative critically ill patients who were randomized to ketamine or saline infusions. The mean cumulative morphine consumption at 48 hours was significantly lower in the ketamine group (58 ± 35 mg) compared to the morphine-only group (80 ± 37 mg; P < .05). Other trials showed the potential safety of ketamine in terms of cerebral hemodynamics in patients with traumatic brain injury, improved gastrointestinal motility, and decreased vasopressor requirements. The observational study and case reports suggest that ketamine is safe and effective and may have a role in patients who are refractory to other therapies. Conclusion: Ketamine use may decrease analgesic consumption in the intensive care unit. Additional trials are needed to further delineate the role of ketamine for analgosedation.
APA, Harvard, Vancouver, ISO, and other styles
48

Pemayun, I. Gusti Agung Gde Putra, and I. Gusti Ngurah Sudisma. "Anestesi Tetes Infus Gravimetrik Ketapol sebagai Alternatif Bius Umum Secara Inhalasi Guna Menjaga Status Teranestesi pada Babi." Jurnal Veteriner 19, no. 1 (July 13, 2018): 126. http://dx.doi.org/10.19087/jveteriner.2018.19.1.126.

Full text
Abstract:
This study aim was to evaluate quality and effectiveness of anaesthesia by using gravimetric infusion anaesthesia with ketamine and propofol (ketafol) on pigs. The quality of anesthesia time, the cardiovascular and respiratory response of anaesthesia were evaluated in twelve male pigs with average body weight 35±5 kg. The pigs were divided into four treatments and each treatment consisted of three pigs as repetition. All the experimental animals were premedicated with atropine 0.03 mg/kg mixed with xylazine 2 mg/kg in one syringe injected intramuscularly. Fifteen minutes after premedicated, the pigs was induced intravenously with ketamine 6 mg/kg and propofol 2 mg/kg. Furthermore, the anesthetized state maintaned with gravimetric, each through infusion with propofol (0.1 mg/kg/minute) (PI-P) , ketamine (0.3 mg/kg/ minute) (PIK), combination propofol-ketamin (0.1 and 0.3 mg/kg/minute) (PI-PK), and inhalation with isoflurane 1-2% (PI-I). The heart rate, pulse, respiratory rate, rectal temperature, blood oxygen saturation (SpO2 ), capillary refill time (CRT) were observed before and after treatment of the anesthetic. Premedication combination of atropin 0.03mg/kg mixed with xylazine 2 mg/kg in one syringe and then induced with ketamine 6 mg/kg and propofol 2 mg/kg showed the average length of anesthesia for 32,33 minutes, longer compared to five other treatments with average length of induction three minutes and average recovery time 20.33 minutes. Maintenance of anesthesia with propofol infusion drops (0.1 mg/kg/minute), or propofolketamine infusion drops (0.3 and 0.1 mg/kg/minute) showed safer anesthesia, because they did not make drastic change of heart rate, respiratory rate, rectal temperature, oxygen saturation, pulse, and CRT value in the experimental pigs. The conclusion of this study is the methode of gravimetric intravenous infusion by using ketamine and propofol (ketafol) can be used to maintain anesthesia as an alternative general inhalation anaesthesia in pigs.
APA, Harvard, Vancouver, ISO, and other styles
49

Zheng, Wei, Yan-Ling Zhou, Cheng-Yu Wang, Xiao-Feng Lan, Bin Zhang, Su-Miao Zhou, Su Yan, and Yu-Ping Ning. "Plasma BDNF concentrations and the antidepressant effects of six ketamine infusions in unipolar and bipolar depression." PeerJ 9 (March 29, 2021): e10989. http://dx.doi.org/10.7717/peerj.10989.

Full text
Abstract:
Objectives Accumulating evidence has implicated that brain derived neurotrophic factor (BDNF) is thought to be involved in the pathophysiology of depression, but its correlation with ketamine’s antidepressant efficacy focusing on Chinese individuals with depression is not known. This study was aim to determine the correlation of plasma BDNF (pBDNF) concentrations and ketamine’s antidepressant efficacy. Methods Ninety-four individuals with depression received six intravenous infusions ketamine (0.5 mg/kg). Remission and response were defined as Montgomery-Asberg Depression Rating Scale (MADRS) scores less than 10 and a reduction of 50% or more in MADRS scores, respectively. Plasma was collected at baseline and at 24 h and 2 weeks after completing six ketamine infusions (baseline, 13 d and 26 d). Results A significant improvement in MADRS scores and pBDNF concentrations was found after completing six ketamine infusions compared to baseline (all ps < 0.05). Higher baseline pBDNF concentrations were found in ketamine responders/remitters (11.0 ± 6.2/10.1 ± 5.8 ng/ml) than nonresponders/nonremitters (8.0 ± 5.5/9.2 ± 6.4 ng/ml) (all ps < 0.05). Baseline pBDNF concentrations were correlated with MADRS scores at 13 d (t = − 2.011, p = 0.047) or 26 d (t = − 2.398, p = 0.019) in depressed patients (all ps < 0.05). Subgroup analyses found similar results in individuals suffering from treatment refractory depression. Conclusion This preliminary study suggests that baseline pBDNF concentrations appeared to be correlated with ketamine’s antidepressant efficacy in Chinese patients with depression.
APA, Harvard, Vancouver, ISO, and other styles
50

Zaifada, A. U. "Effects of Combinations of Medetomidine-Diazepam-Ketamine and Medetomidine-Ketamine Anesthesia on Haematology and Serum Biochemistry in Dogs." Journal of Veterinary and Biomedical Sciences 4, no. 1 (June 28, 2022): 94–101. http://dx.doi.org/10.36108/jvbs/2202.40.0111.

Full text
Abstract:
This study was aimed at evaluating the effects of combinations of medetomidine-diazepamketamine and medetomidine-ketamine anesthesia haematology and serum chemistry in dogs. Ten (10) apparently healthy local dogs comprising of five (5) females and five (5) males with Mean ± SD body weight of 20.4 ± 2.93 kg sought from Maiduguri and environs and randomly allocated into two groups A (n=5), B (n=5) were used to conduct the experiment for this study. Total Intravenous anesthesia (TIVA) was induced in all the dogs. Dogs in group A were premedicated with an intravenous injection of 0.005 mg/kg Medetomidine, followed by an intravenous injection of 0.25 mg/kg Diazepam and 4 mg/kg Ketamine combination 3 – 5 minutes later. Meanwhile group B dogs were given an intravenous injection of Medetomidine (0.005 mg/kg) and Ketamine (5 mg/kg) combination. Blood sample was collected for haematological analysis while serum was collected to assay Total protein (TP), Alanine Aminotransferase (ALT), Blood urea nitrogen (BUN) and Creatinine (Cr) using standard laboratory protocols. There was no significant difference (p>0.05) in the measured haematological indices, ALT, BUN, Cr and TP in both groups A and B throughout the study. However, there was non-significant decrease in ALT, BUN, Cr and TP in both groups up to 30 minutes post administration of the drug combinations but return to almost baseline values at 60 minutes post anaesthesia. The results of this study showed that intravenous administration of Medetomidine-Diazepam-Ketamine and Medetomidine-Ketamine combinations have no significant effects on haematological indices and serum chemistry profile of dogs. Therefore, these anaesthetic combinations can be prescribed for clinical use and procedures in dogs.
APA, Harvard, Vancouver, ISO, and other styles
We offer discounts on all premium plans for authors whose works are included in thematic literature selections. Contact us to get a unique promo code!

To the bibliography