Your activity: 20721 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: [email protected]

Procedural sedation in adults outside the operating room

Procedural sedation in adults outside the operating room
Author:
Robert L Frank, MD, FACEP
Section Editor:
Allan B Wolfson, MD
Deputy Editor:
Michael Ganetsky, MD
Literature review current through: Feb 2022. | This topic last updated: Aug 17, 2021.

INTRODUCTION — The practice of acute care medicine often requires the performance of procedures that can cause pain and anxiety. Procedural sedation reduces the discomfort, apprehension, and potential unpleasant memories associated with such procedures and facilitates performance of the procedure.

The practice of procedural sedation in adults, including monitoring and medications, will be reviewed here. Procedural sedation in children and the sedation required for lengthier procedures, such as colonoscopy, are discussed separately. (See "Procedural sedation in children outside of the operating room" and "Gastrointestinal endoscopy in adults: Procedural sedation administered by endoscopists".)

DEFINITIONS — Procedural sedation involves the use of short-acting analgesic and sedative medications to enable clinicians to perform procedures effectively, while monitoring the patient closely for potential adverse effects. This process was previously (and inappropriately) termed "conscious sedation," but because effective sedation often alters consciousness, the preferred term is now "procedural sedation and analgesia" (PSA) [1].

The International Committee for the Advancement of Procedural Sedation defines it as follows [2]:

"The practice of procedural sedation is the administration of one or more pharmacological agents to facilitate a diagnostic or therapeutic procedure while targeting a state during which airway patency, spontaneous respiration, protective airway reflexes, and hemodynamic stability are preserved, while alleviating anxiety and pain."

The Committee further notes that procedural sedation may occur with concurrent blunting of airway reflexes with local anesthesia and that sedation states within the purview of the definition include minimal sedation, moderate sedation, dissociative sedation, and deep sedation. These are defined below. It further notes that procedural sedation differs from general anesthesia, "which targets an unarousable state in which airway intervention is often required and spontaneous ventilation is frequently inadequate."

The practice of providing sedation, once primarily the domain of anesthesia practitioners, is now routinely performed by other specialists, such as emergency clinicians, critical care specialists, and various nurse specialists [3].

Recognizing that PSA exists along a spectrum, the Joint Commission on Accreditation of Healthcare Organizations in the United States has attempted to define the levels of sedation, which range from minimal sedation to general anesthesia [4-6]. Common terms include the following:

Analgesia – Relief of pain without intentionally producing a sedated state. Altered mental status may occur as a secondary effect of medications administered for analgesia.

Minimal sedation – The patient responds normally to verbal commands. Cognitive function and coordination may be impaired, but ventilatory and cardiovascular functions are unaffected.

Moderate sedation and analgesia – The patient responds purposefully to verbal commands alone or when accompanied by light touch. Protective airway reflexes and adequate ventilation are maintained without intervention. Cardiovascular function remains stable.

Deep sedation and analgesia – The patient cannot be easily aroused but responds purposefully to noxious stimulation. Assistance may be needed to ensure the airway is protected and adequate ventilation maintained. Cardiovascular function is usually stable.

General anesthesia – The patient cannot be aroused and often requires assistance to protect the airway and maintain ventilation. Cardiovascular function may be impaired.

Dissociative sedation – Dissociative sedation is a trance-like cataleptic state in which the patient experiences profound analgesia and amnesia but retains airway protective reflexes, spontaneous respirations, and cardiopulmonary stability [7]. Ketamine is the pharmacologic agent used for procedural sedation that produces this state. (See 'Ketamine' below.)

Sedation exists on a continuum, and it is difficult to divide it into discrete clinical stages, the definitions above notwithstanding [6]. Moreover, many sedatives can cause rapid changes in the depth of sedation. Dissociative sedation stands apart from the continuum of sedation due to its unique characteristics.

INDICATIONS — There are no absolute indications for the performance of procedural sedation and analgesia (PSA). PSA may be used for any procedure in which a patient's pain or anxiety may be excessive and may impede performance. PSA is often useful for procedures where deep relaxation facilitates performance (eg, closed reduction of a dislocated joint). Common procedures in which PSA may be beneficial include electrical cardioversion, closed joint reduction, complicated laceration repair, abscess incision and drainage, and lumbar puncture.

CONTRAINDICATIONS AND PRECAUTIONS

General considerations — There are no absolute contraindications to procedural sedation and analgesia (PSA). Relative contraindications may include: older age, significant medical comorbidities, and signs of a difficult airway. Whether the patient recently ate should be considered before performing PSA, although this does not appear to have a major impact on aspiration risk [8,9]. (See 'Aspiration risk' below.)

Several factors must be considered before proceeding with PSA. First, the clinician and the patient must agree that the potential benefit of PSA outweighs the risks. Risk depends upon the patient and the procedure.

There is no specific age above which PSA may not be performed. Nevertheless, the elderly have higher rates of adverse events [10]. This may be due to an increased sensitivity to sedative drugs, medication interactions, and higher peak serum levels of administered drugs [11-13].

Patients with major comorbid medical conditions are at increased risk for adverse events with PSA. This correlates with an ASA physical status classification of Class III or greater (table 1) [11,13]. Important comorbidities are those that increase patient susceptibility to the cardiorespiratory depressant effects of sedatives. They include heart failure, chronic obstructive pulmonary disease, neuromuscular disease, dehydration, and anemia. Unfortunately, there is no evidence that alternative approaches (eg, monitored anesthesia care or general anesthesia in the operating room) are safer for patients at increased risk from PSA.

To reduce the risk of adverse events in older adults and patients with major comorbid disease, we suggest a more conservative approach to PSA medications, including:

Giving a lower starting dose

Using slower rates of administration

Repeated dosing of medications at less frequent intervals

PSA is relatively contraindicated in patients who are likely to be difficult to ventilate or oxygenate should respiratory difficulties arise while the patient is sedated. Alternatives to PSA may be preferable if signs suggesting a difficult airway are identified. (See "Approach to the anatomically difficult airway in adults outside the operating room".)

No data clearly demonstrate that a longer duration of PSA correlates with an increase in adverse outcomes. Nevertheless, common sense suggests that it is preferable to sedate patients for the shortest period necessary to perform the procedure.

Aspiration risk — Clinically significant aspiration of gastric contents during procedural sedation is extremely rare. A systematic review identified only nine deaths, eight of which occurred during upper gastrointestinal endoscopy; none occurred in healthy children or adults [14]. Available evidence suggests that fasting prior to procedural sedation is unnecessary for preventing pulmonary aspiration of gastric contents in most cases [15,16].

The American College of Emergency Physicians (ACEP), in the 2018 practice guideline "Unscheduled Procedural Sedation: A Multidisciplinary Consensus Approach," states: "Providers of unscheduled procedural sedation should assess the timing and nature of recent oral intake. The urgency of the procedure will dictate the necessity of providing sedation without delay regardless of the fasting status" [17]. Thus, recent food intake is not a contraindication for administering procedural sedation and analgesia but should be considered when choosing the timing and target of sedation. We suggest the following approach to reducing aspiration risk:

Carefully consider the risks and benefits of performing the procedure immediately. Although there is no proof that longer fasting times reduce aspiration risk, it may be reasonable to wait if the patient's stomach is full and the procedure is not a true emergency [18]. This is particularly true when a potentially difficult airway or an increased risk for aspiration exists, as with the following circumstances [17,19]:

Conditions predisposing to esophageal reflux (eg, bowel obstruction, hiatal hernia)

Extremes of age (<6 months or >70 years old)

Severe systemic disease (ASA class III or greater)

Obstructive sleep apnea

Obesity    

Other concerning conditions (eg, depressed mental status)

Avoid deep sedation. No evidence clearly demonstrates that deeper levels of sedation increase the risk of aspiration [19]. Nevertheless, lighter sedation may permit the patient to maintain protective airway reflexes, which reduces risk.

Consider using ketamine for sedation in cases where delay is not possible and the risk of aspiration is elevated, as ketamine maintains protective airway reflexes [20,21].

Avoid the administration of preprocedural antacids or promotility medications. These medications have not been shown to reduce aspiration risk [22].

In some cases, it may be best to perform the procedure under general anesthesia in the operating room, although this approach has not been proven to reduce the risk of aspiration [23,24].

Clinically significant aspiration of gastric contents during anesthesia or PSA is a rare, though much feared, complication [8,9]. Patients undergoing emergency procedures requiring sedation are thought to be at increased risk of aspiration because their stomachs are often full, and the procedure cannot be delayed. Aspiration frequently does not cause harm. However, aspirated gastric contents above a critical volume and acidity can cause severe respiratory and systemic consequences [25].

Evidence suggests no direct relationship between any specific period of preprocedural fasting and gastric acid volumes and pH [7,13,19,22,23,26,27]. Also, episodes of emesis, apnea, or hypoxemia have not been shown to be reduced by periods of preprocedure fasting [28,29]. In addition, endotracheal intubation may not protect the patient from aspiration. Aspiration can occur despite the presence of an endotracheal tube, and airway manipulation involved in performing intubation may increase the risk of aspiration [19,24,26,30-32]. In the emergency setting, it is also not often feasible to delay procedural sedation for emergent procedures. Taking this evidence into consideration, the latest ASA guidelines published in 2018 state: "In urgent or emergent situations where complete gastric emptying is not possible, do not delay moderate procedural sedation based on fasting time alone" [33]. These guidelines make no recommendations for patients requiring deep sedation.

For elective procedures in adults, the 2018 ASA guidelines recommend no oral intake (NPO or nil per os) for a minimum of two hours after consuming clear liquids and a minimum of six hours after consuming solid food or milk.

PERFORMING PROCEDURAL SEDATION

Informed consent — Before performing procedural sedation and analgesia (PSA), the clinician must discuss the risks, benefits, and alternatives of the procedure and the planned sedation with the patient and answer any questions. A printed informed consent form may be used. Implied consent is acceptable in some cases where the patient is unable to provide explicit consent due to severe pain or altered mental status [34].

Prerequisites and personnel — Although previously the domain of anesthesia practitioners, PSA is performed safely by other clinicians, including emergency and critical care physicians and nurse specialists [35]. Clinicians providing PSA should have in-depth knowledge of the relevant drugs, including their mechanism of action, doses, side effects, and reversal agents. Such clinicians must also be well versed in advanced cardiovascular life support, including airway management. (See "Advanced cardiac life support (ACLS) in adults".)

The number of clinicians needed to perform PSA and the procedure safely may vary according to the patient and the procedure. In most cases, one clinician performs the procedure while another (usually a nurse) administers the sedative agents and monitors and records the patient's vital signs and clinical status. Whenever possible, we suggest that this minimum standard be met [15,33].

It remains controversial whether an additional clinician (separate from the clinician performing the procedure) who is skilled in deep sedation administration and airway management should be present [13,34]. Guidelines from the American Society of Anesthesiologists (ASA) call for an assistant(s) other than the practitioner performing the procedure who has "advanced life support skills" and can monitor the patient, obtain intravenous access, and recognize and treat complications related to the performance of procedural sedation [4,33]. Large case series of propofol sedation administered by nurses for ambulatory procedures demonstrate that serious adverse events can occur [36]. However, one prospective observational study of over 1000 procedural sedations found that PSA performed by a single emergency clinician (EP) had comparably low complication and high success rates when compared with PSA performed with both an EP and a nurse present [37].

Equipment — All equipment necessary to perform the procedure and manage the airway should be available at the bedside during the performance of PSA. Such equipment includes suction to manage vomiting or oral secretions, airway adjuncts, such as a bag-valve mask and oral and nasal airways, and equipment to perform endotracheal intubation. Intravenous access should be established. Resuscitation medications, including advanced cardiac life support medications and reversal agents (ie, naloxone and flumazenil) should be available. Appropriate monitors should be in place. (See 'Monitoring and preoxygenation' below.)

Monitoring and preoxygenation — Proper monitoring of patients during the performance of PSA is crucial. The patient's blood pressure, heart rate, and respiratory rate should be measured at frequent, regular intervals; the oxygen saturation (SpO2), end-tidal carbon dioxide (EtCO2) level, and cardiac rhythm should be monitored continuously [13].

The patient's response to medications and the procedure must also be closely monitored during PSA. The patient's level of alertness, depth of respiration, and response to painful stimuli (eg, fracture reduction) are all important factors in determining subsequent medication doses. Sedation scales, such as the Richmond Agitation Sedation Scale and the Ramsay Score, have not been adequately studied in the setting of PSA. They may be more useful in determining the appropriate titration of sedatives during long-term procedures (eg, mechanical ventilation).

Supplemental oxygen is often recommended during PSA to maintain oxygen reserves and prevent hypoxemia caused by hypoventilation [4,33]. However, there is little evidence to show that this is beneficial and some researchers question the practice.

The best evidence supporting the use of oxygen is a double blind, randomized trial of adults undergoing PSA with propofol in which episodes of hypoxia (SpO2 <93 percent) lasting longer than 15 seconds occurred significantly more often (41 percent) among the 58 patients given compressed air by face mask compared to the 59 patients given high flow oxygen (19 percent) using the same delivery system (difference 23 percent; 95 percent CI 6-38 percent). [38]. However, the clinical significance of such transient episodes of hypoxia remains debatable.

Several observational studies have found that supplemental oxygen at lower concentrations does not reliably prevent hypoxemia during PSA [39,40], and delays the detection of respiratory depression in patients without EtCO2 monitors, since SpO2 levels may not fall until a prolonged period of hypoventilation or apnea has occurred [41,42]. Other data, however, have shown reductions in procedural hypoxemia when patients are given oxygen [39,43]. Recommendations from the ASA guidelines on procedural sedation recommend the use of supplemental oxygen unless contraindicated, although there are no data to suggest the best delivery system [33]. We suggest that high flow oxygen by face mask be given to patients undergoing PSA because it may reduce the likelihood of hypoxic episodes, particularly with prolonged procedures, is easy to perform, and is highly unlikely to cause harm.

However, among patients not provided with supplemental oxygen, respiratory difficulty during PSA can manifest earlier as hypoxia, as detected by pulse oximetry, rather than as hypoventilation, as determined by EtCO2 monitors, according to a small nested randomized trial [44]. Therefore, close attention to pulse oximetry readings remains critical when monitoring patients' breathing room air during PSA.

Although evidence is limited, we recommend pulse oximetry and EtCO2 monitoring for all patients undergoing PSA [45]. EtCO2 measurements correlate closely with arterial CO2 and provide an early sign of hypoventilation or apnea, especially if supplemental oxygen is used [15,41,46,47]. EtCO2 is discussed in detail separately. (See "Carbon dioxide monitoring (capnography)".)

While intuitively it makes sense that early detection of hypoxemia and hypoventilation is beneficial, there is no data to suggest that brief episodes of either have a negative impact on patient outcome, especially if recognized quickly and treated appropriately [34,48].

Bispectral analysis monitoring (BIS), a technology developed to monitor the level of general anesthesia, does not appear to be useful for monitoring the depth of procedural sedation [49-51]. BIS measurements do not appear to correlate well with clinical sedation and have poor reproducibility.

Considerations in obese adults — Adjustments in management and drug dosing are often necessary when providing procedural sedation to obese adults due to physiologic changes and associated health problems, such as sleep apnea and restrictive lung disease [52]. These conditions may predispose to hypoxemia and difficulties with ventilation and other aspects of airway management [53,54]. The physiologic changes and medical conditions associated with obesity, and important aspects of airway management in obese adults, are discussed separately. (See "Overweight and obesity in adults: Health consequences" and "Emergency airway management in the morbidly obese patient".)

Procedural sedation in obese patients is associated with a more frequent need for airway maneuvers (eg, bag-mask ventilation) and more frequent, albeit brief, episodes of hypoxemia, but does not appear to increase the incidence of serious adverse outcomes or of premature termination of the procedure [55]. However, obesity may affect the choice of sedative agent and dosing, as pharmacokinetics and pharmacodynamics can be changed by the increased volume of distribution, altered drug clearance, and increased cardiac output seen with obesity.

Drug dosing — Adjustments in the dosing of drugs used for procedural sedation are described below. As a general rule of thumb, dosing should be based on ideal or lean bodyweight to avoid oversedation.

Propofol – A reasonable strategy when performing PSA with propofol in the obese is to give an initial dose based on the patient's ideal body weight and then give additional titrated doses as needed to achieve the desired level of sedation. Propofol's pharmacokinetics are highly dependent on cardiac output, which is elevated in obese patients. Propofol is also highly lipophilic and thus has a greater volume of distribution in obese patients. Both these factors can lead to more rapid clearance and a shorter duration of effect.

While some have suggested that dosing be based on total body weight (TBW) [56,57], others have found that the relationship between weight and pharmacokinetic variables is non-linear, and dosing based on TBW may result in doses of propofol that are excessive for PSA [58].  

Ketamine – We suggest that ideal body weight be used to determine the initial dosing of ketamine for PSA, with additional titrated doses provided as needed. Little evidence is available to determine the best dosing regimen for ketamine when used for PSA in obese patients. While some have suggested dosing based on TBW, these higher doses may increase the risk of side effects and oversedation [56]. Others have suggested using ideal body weight or adjusted body weight, as we do [58].  

Etomidate – Etomidate has pharmacokinetic and pharmacodynamic properties similar to propofol. Thus, we suggest that dosing be based on ideal body weight, with additional titrated doses given as needed [59].

Dexmedetomidine – We suggest that lean or ideal body weight, not TBW, be used to determine the dosing of dexmedetomidine for PSA [58,60]. Dexmedetomidine's pharmacodynamics and pharmacokinetics differ in the obese and lower doses are needed to reach a given plasma concentration and level of sedation.

Opioids – We suggest that lean body weight be used to determine the dosing of drugs such as fentanyl, alfentanil, and remifentanil for procedural sedation. Doses based on TBW increase the risk for respiratory depression, hypotension, and bradycardia [59].

Benzodiazepines – Benzodiazepines (eg, diazepam, midazolam) are seldom used as single agents for procedural sedation, but TBW may be used to calculate initial intravenous doses of a benzodiazepine given for this purpose [56]. However, the half-life and volume of distribution of benzodiazepines increases with body weight, and so there is the potential for the drug and metabolites to accumulate with additional doses, potentially causing adverse effects such as oversedation and respiratory depression. Thus, regardless of whether benzodiazepines are used alone or in combination with other medications, it is safer to use ideal body weight to determine any subsequent doses [58].

Sedative-hypnotics – Medications such as thiopental and methohexital are used infrequently for PSA, but when used, we suggest using ideal body weight to calculate the initial dose, with additional titrated doses given as needed to achieve adequate sedation [61]. The increased cardiac output seen in obese patients can cause more rapid redistribution and thus more rapid awakening after a single bolus dose of these medications [59].

Considerations in pregnancy — Modifications of procedural sedation guidelines recommended for pregnant women include:

Pre-procedural administration of medication to improve gastroesophageal sphincter tone and reduce gastric volume (eg, metoclopramide) and decrease stomach acidity (eg, H2 antagonists, sodium citrate) may reduce the risk of vomiting and aspiration and is unlikely to cause harm. Aspiration risk is discussed separately. (See 'Aspiration risk' above.)

Pre-procedural hydration and left lateral displacement of the uterus (in the late second and the third trimester) helps to reduce the risk of hypotension, uteroplacental insufficiency, and resultant fetal hypoxemia. Fetal monitoring is not required but should be considered for women in the third trimester. (See "Nonstress test and contraction stress test".)

Oxygen by face-mask is administered because of the risk of sedation-related maternal desaturation (primarily due to decreased functional residual capacity). (See "Maternal adaptations to pregnancy: Cardiovascular and hemodynamic changes" and "Maternal adaptations to pregnancy: Dyspnea and other physiologic respiratory changes", section on 'Physiologic cardiopulmonary changes in pregnancy'.)

COMPLICATIONS — Serious complications attributable to PSA rarely occur [15,62,63]. According to a systematic review of 55 studies including 9652 cases of PSA performed in the emergency department, the rate of severe adverse events requiring an emergency intervention is exceedingly low [62]. Adverse outcomes may include respiratory depression with hypoxia or hypercarbia, cardiovascular instability, vomiting and aspiration, emergence reactions, and inadequate sedation preventing completion of the procedure [11]. However, significant respiratory compromise, the most concerning potential complication, develops in well less than 1 percent of cases. Among the studies included, the review identified one case of aspiration in 2370 sedations (1.2 per 1000), one case of laryngospasm in 883 sedations (4.2 per 1000), and two intubations in 3636 sedations (1.6 per 1000).

Multiple studies of pediatric patients have found that PSA is safe, with no deaths reported and few serious consequences [36,64-66]. One observational case series of 640 patients undergoing PSA reported a 15 percent combined adverse event rate (adverse event and failure to perform procedure) [10]. In this study, factors associated with adverse events included older age, the presence of multiple medical conditions (ie, higher ASA class), and the performance of esophagoscopy or cardioversion.

Many complications can be prevented through appropriate selection of patients, proper use of sedative medication, and careful monitoring of sedation. Particular attention should be paid to patients in whom oxygenation and ventilation may be difficult, should the need for airway management arise. Such patients may not be appropriate candidates for PSA. (See 'Contraindications and precautions' above.)

Nearly all of the sedative agents used for PSA can cause dose-dependent respiratory depression [11]. Therefore, respiratory complications are the most common adverse events. Oxygen desaturation develops in up to 11 percent of adults who receive PSA with either propofol or etomidate and are given supplemental oxygen [13]. Rates are slightly higher if supplemental oxygen is not used. Oxygen desaturation can be minimized by cautious, unhurried medication administration [11].

Hypoventilation and apnea may occur but are usually short-lived due to the brief duration of the drugs used for PSA. These complications nearly always resolve with patient stimulation, supplemental oxygen, positioning of the airway, or brief ventilatory support using a bag-valve mask. Treatment with the reversal agent naloxone or flumazenil may be necessary with more severe or prolonged respiratory depression during PSA using opioids or benzodiazepines. The use of naloxone and flumazenil are reviewed separately. (See "Acute opioid intoxication in adults", section on 'Basic measures and antidotal therapy' and "Benzodiazepine poisoning and withdrawal", section on 'Antidote (flumazenil)'.)

Significant hypotension and bradycardia seldom occur but may develop in patients with significant cardiac morbidity and those taking cardio-depressant medications (eg, beta blockers) [11,67-69]. These problems are usually transient and resolve without intervention. Hemodynamically neutral sedatives (eg, etomidate) may be preferable for patients at risk from changes in blood pressure or heart rate.

Respiratory and cardiovascular side effects can be minimized by titrating the medication in small incremental doses, titrating to desired effect, and permitting the peak effect to occur before giving additional medication [33].

Nausea and vomiting occur in about 5 percent of patients undergoing PSA, although rates may be higher when opioids are used [70-73]. Antiemetics may be used as needed. There is little evidence about the prophylactic use of antiemetics or which agent is preferable. In one randomized trial involving PSA with ketamine, 16 of 127 children managed without antiemetics experienced post-procedural vomiting versus 6 of 128 children pretreated with ondansetron, a 7.9 percent reduction in absolute risk (95% CI 1.1-14.7) [74].

MEDICATIONS — Procedural sedation and analgesia (PSA) typically involves the intravenous (IV) administration of sedative or dissociative agents, sometimes in combination with short-acting opioids (table 2) [34]. Ideal drugs for PSA have a rapid onset and short duration of action, maintain hemodynamic stability, and do not cause major side effects [3]. Several medications are commonly used and no single drug is ideal for all situations. Dosing modifications when performing PSA in obese adults are reviewed above. (See 'Considerations in obese adults' above.)

Propofol — Propofol is a phenol derivative that has been shown to provide effective PSA for emergent procedures [3,63]. Propofol is highly lipophilic and therefore crosses the blood-brain barrier rapidly. The drug takes effect within approximately 40 seconds, and its duration of action is approximately six minutes [13,75]. Propofol acts as a sedative and amnestic but provides no analgesia. In addition to its use for PSA, propofol is used for long-term sedation in critically ill patients, which is discussed separately. (See "Sedative-analgesic medications in critically ill adults: Properties, dosage regimens, and adverse effects", section on 'Propofol'.)

Propofol can induce deep sedation rapidly and must be given with careful attention to dosing and monitoring. For PSA in adults, propofol is given by slow injection in an initial loading dose of 0.5 to 1 mg/kg IV, followed by doses of 0.25 to 0.5 mg/kg IV every one to three minutes as necessary until the appropriate level of sedation is achieved [63]. One reasonable approach to administration is to give 20 mg every 10 seconds (eg, a 50 mg dose would be given over 25 seconds), although there is no direct evidence demonstrating improved efficacy or safety using this regimen. (See 'Monitoring and preoxygenation' above.)

The pharmacokinetics of propofol appear to be unchanged in patients with impaired kidney or liver function. However, plasma levels appear to be increased in older adult patients, which can lead to prolonged sedation and more pronounced cardiorespiratory depression. Patients over 55 are particularly sensitive [13]. The manufacturer recommends that doses in older adults be reduced by 20 percent and that the drug be given more slowly (over three to five minutes) [76]. Reductions from 20 to 60 percent of the dose used in a healthy young adult are reasonable. (See 'Older adult patients' below.)

Although the manufacturer lists egg or soybean allergies as contraindications to the use of propofol, significant allergic reactions to the newer preparation of the drug appear to be rare. (See "Perioperative anaphylaxis: Clinical manifestations, etiology, and management", section on 'Hypnotic induction agents'.)

Potential side effects include hypotension (due to myocardial depression) and respiratory depression. These side effects generally resolve quickly and uneventfully because of the brief duration of action. However, hypotension can produce complications in patients with severe medical problems (eg, sepsis, cardiac dysfunction) or hypovolemia [13].

Respiratory depression usually manifests as a mild oxygen desaturation. Coadministration with other sedatives or analgesics (eg, fentanyl) can exacerbate respiratory problems. Episodes of hypoxia are generally uneventful and successfully treated with supplemental oxygen and patient stimulation, and less commonly require short periods of assisted ventilation with a bag-valve mask. There are no reports of endotracheal intubation due to propofol-induced respiratory depression during PSA [13].

Propofol provides no analgesia and can cause pain during injection through an intravenous catheter. According to a systematic review of 177 randomized controlled trials involving 25,260 adult patients, the following measures are significantly effective at reducing pain caused by propofol injection [77]:

Injecting into an antecubital vein rather than a hand vein (relative risk [RR] 0.14; 95% CI 0.07-0.30)

Lidocaine pretreatment while occluding the vein (RR 0.29; 95% CI 0.22-0.38)

Pretreatment with lidocaine-propofol admixture (RR 0.40; 95% CI 0.33-0.48)

Lidocaine pretreatment without vein occlusion (RR 0.47; 95% CI 0.40-0.56)

Opioid pretreatment (RR 0.49; 95% CI 0.41-0.59)

Ketamine pretreatment (RR 0.52; 95% CI 0.46-0.57)

Pain during injection can also be reduced by using larger catheters placed in larger veins.

Pretreatment with any of several analgesics can reduce the pain caused by propofol. One effective approach is to place a tourniquet on the arm proximal to the injection site and then give 0.5 mg/kg of lidocaine IV 30 to 120 seconds before propofol is injected [75].

Another approach is pretreatment with short-acting opioids (eg, fentanyl). Unfortunately, the addition of opioids increases the likelihood of respiratory complications. As an example, fentanyl at a dose of 1.5 mcg/kg increases the risk of respiratory depression. To reduce this risk, smaller doses of fentanyl should be used. The best dose to provide adequate analgesia with minimal respiratory risk has yet to be identified. We suggest that no single dose of fentanyl exceed 0.5 mcg/kg when given with propofol and that the total amount of fentanyl be kept to the minimum required for effective analgesia.

Ketamine is an alternative to opioids and lidocaine for reducing the pain of injection. In a randomized trial performed in patients undergoing elective surgery, ketamine given in doses of 100 mcg/kg (around 10 mg) immediately before injection of propofol was found to be safe and effective in alleviating the pain from propofol injection [78].

It may not be necessary to combine propofol with a short-acting opioid if the patient's pain is adequately treated prior to the procedure. This approach is supported by an unblinded randomized trial in which patients given PSA consisting of propofol alone had pain levels identical to those treated with both propofol and alfentanil [79]. For both treatment groups, intravenous morphine was given at least 20 minutes prior to the procedure to all patients experiencing pain until their symptoms were adequately relieved. Of note, patients given both alfentanil and propofol required stimulation to induce respiration more often than those given only propofol.

Another alternative to pretreatment with short-acting opioids is to use sub-dissociative doses of ketamine given 0.1 to 0.3 mg/kg [80,81]. Ketamine appears to provide comparable analgesia with less risk of respiratory depression. (See 'Ketamine versus short-acting opioids for analgesia' below.)

In some instances, patients experience little or no pain before or after a procedure (eg, lumbar puncture, cardioversion) and little if any analgesia may be necessary to supplement propofol. In addition, some procedures lead to a substantial reduction in pain (eg, dislocation reduction), decreasing the need for analgesics as part of PSA.

Multiple randomized trials and prospective observational studies have found propofol to be safe and effective for PSA in the emergency department [13,82,83]. Even in the contentious area of nurse-administered propofol sedation, where typically the only physician present is performing the procedure (eg, outpatient endoscopy), there is a large body of evidence documenting the safety and efficacy of propofol [84,85]. Studies comparing propofol to alternative medications for PSA are limited [86].

Despite the evidence above, not all clinicians who are qualified and wish to administer propofol for PSA are allowed to do so. Reasons for this are complex and beyond the scope of this discussion but may include misunderstandings about patient safety as well as political and economic factors [87,88]. The manufacturer's label states: "only those persons trained in the administration of general anesthesia should administer the drug," and goes on to state: "only those persons not involved in the conduct of the surgical/diagnostic procedure should administer the drug" [89]. In the United States, these statements have led some nursing organizations to question whether nurses other than certified registered nurse anesthetists should administer propofol.

Etomidate — Etomidate, an imidazole derivative, is a sedative that is commonly used for PSA. The use of etomidate for rapid sequence intubation is discussed elsewhere. (See "Induction agents for rapid sequence intubation in adults outside the operating room".)

For PSA in adults, etomidate is given IV over 30 to 60 seconds in doses of 0.1 to 0.15 mg/kg, less than the dose used for rapid sequence intubation. It can be redosed approximately every three to five minutes as needed. The onset of action of etomidate is almost immediate and its duration of effect is 5 to 15 minutes [90,91].

Etomidate can have more profound and prolonged effects in the elderly and patients with renal or hepatic dysfunction. In such patients, doses in the lower dosing range should be used. An important benefit of etomidate is that it maintains cardiovascular stability.

Etomidate has no analgesic properties and often requires the coadministration of a short-acting opioid, such as fentanyl, which increases the risk of respiratory depression [91]. To reduce this risk, smaller doses of fentanyl should be used. The best dose to provide adequate analgesia with minimal respiratory risk has yet to be identified. We suggest that no single dose of fentanyl exceed 0.5 mcg/kg when given with etomidate and that the total amount of fentanyl be kept to a minimum.

Etomidate causes pain during injection into peripheral veins. Strategies similar to those used for propofol can be used to reduce such pain. (See 'Propofol' above.)

Several randomized trials and prospective observational studies have found that etomidate is an effective sedation agent for PSA and does not cause major complications (eg, respiratory depression requiring endotracheal intubation) [90,92-94]. However, side effects, particularly myoclonus, occur with regularity and procedure success rates may be lower when compared with propofol or ketamine. (See 'Propofol versus etomidate' below.)

Potential side effects of etomidate include myoclonus, respiratory depression, adrenal suppression, and nausea and vomiting. Myoclonus is the most frequently reported side effect. It is thought to be related to subcortical disinhibition and has been reported in up to 80 percent of patients who receive etomidate for PSA [94-97]. The degree of myoclonus may be dose dependent and ranges from mild and transient to severe enough to prevent completion of the procedure [90].

Reports of severe myoclonus associated with PSA are rare. In such cases, we suggest immediate airway support and treatment with midazolam, 1 to 2 mg IV approximately every 60 seconds until myoclonus abates. Alternative benzodiazepines may be used if midazolam is unavailable.

Strategies to prevent myoclonus vary and there is insufficient evidence to support any one approach:

According to one small randomized trial, a pretreatment dose of 0.03 to 0.05 mg/kg of etomidate given 50 seconds before the PSA dose reduces myoclonus [95].

Another small randomized trial in patients undergoing cardioversion found that a small dose of midazolam (0.015 mg/kg) given at the same time as etomidate prevents myoclonus [98].

In another randomized trial, magnesium sulfate administered 90 seconds prior to etomidate was found to reduce myoclonus [99].

According to a systematic review of etomidate for PSA, respiratory depression occurs in approximately 10 percent of cases [92]. In this review, respiratory depression was defined as a fall in oxygen saturation below 90 percent or apnea. No serious complications occurred as a result and respiratory depression resolved quickly without major interventions in the great majority of cases. Nevertheless, clinicians must be prepared to support the patient's airway and breathing in the event of respiratory compromise, as is true whenever PSA is performed.

When given by continuous infusion, etomidate causes adrenal insufficiency. In addition, reductions in plasma cortisol concentrations have been reported in patients receiving a single induction dose of etomidate [100,101]. However, the clinical significance of transient reductions in cortisol in patients undergoing PSA with etomidate remains unclear. Most such patients are relatively healthy and receive a single sedating dose. In such patients, complications related to adrenal suppression have not been reported.

Benzodiazepines (midazolam) — Benzodiazepines are commonly used for minimal sedation (anxiolysis) but less often for deeper sedation due to the superior effectiveness of the ultrashort-acting agents propofol and etomidate. Benzodiazepines produce anxiolysis and amnesia but have no analgesic properties.

Midazolam is the benzodiazepine used most often for PSA. Because it is lipophilic, midazolam penetrates the blood-brain barrier quickly. Midazolam can be used alone for anxiolysis or in combination with short-acting opioids (eg, fentanyl) for deeper levels of sedation and analgesia. Its time of onset is two to five minutes, and its duration of action is 30 to 60 minutes [102,103].

Midazolam is usually given IV over one to two minutes in doses of 0.02 to 0.03 mg/kg. Often in adults, midazolam is given 0.5 or 1 mg at a time and titrated to effect. No single dose should exceed 2.5 mg. Repeat doses may be given every two to five minutes as necessary.

With repeated doses, midazolam accumulates in adipose tissue, which can significantly prolong sedation [102]. The elderly, obese, and those with renal or hepatic disease are at greater risk of prolonged sedation. In such patients, the use of lower doses, longer dosing intervals, and smaller total amounts reduces risk.

The amount of midazolam necessary for adequate sedation varies based upon many factors, including patient size and age, medication tolerance, comorbidities, and the duration of the procedure. In most cases, PSA can be performed using no more than 5 mg of midazolam. For longer procedures likely to require multiple doses of a sedative, ultrashort-acting agents (eg, propofol) may be preferable. (See 'Propofol' above.)

Compared with ultrashort-acting agents, midazolam has a longer duration of action that makes it better suited for anxiolysis than for PSA [96]. Midazolam can cause respiratory depression in high doses or when given concomitantly with other sedatives or narcotics. For anxiolysis, a single dose of 0.02 mg/kg (approximately 1 to 2 mg) is usually sufficient.

Other benzodiazepines, such as lorazepam and diazepam, are less suited for PSA due to their relatively prolonged onset and duration of action. They also have more side effects and inferior amnestic properties compared with midazolam [103,104].

Short-acting opioids — Opioids are often given alone or in combination with sedatives for PSA. Short-acting agents, such as fentanyl, alfentanil, and remifentanil, are used.

Fentanyl is a synthetic opioid that was frequently used in combination with midazolam to provide analgesia during PSA before propofol and etomidate became widely available. It has 75 to 125 times the potency of morphine, a rapid onset of action (two to three minutes), and a short duration of effect (30 to 60 minutes) but has no amnestic properties [103].

Fentanyl is usually given by slow IV push in doses of 0.5 to 1 mcg/kg every two minutes until an appropriate level of sedation and analgesia is achieved [103]. The maximum total dose is generally 5 mcg/kg or approximately 250 mcg, but higher doses may be needed in some instances.

Fentanyl rarely causes hypotension and does not stimulate histamine release. Its primary side effect is respiratory depression, which is potentiated by the coadministration of sedatives. Patients with renal or hepatic disease and the elderly can experience more prolonged or profound effects. In such patients, the use of lower doses, longer dosing intervals, and smaller total amounts reduces risk.

Remifentanil and alfentanil are opioids similar in structure to fentanyl with a rapid onset and duration of action of approximately five minutes, and both are used for PSA [15,105]. The potency of remifentanil and fentanyl are comparable, but alfentanil is one-fifth to one-tenth as potent. Remifentanil can be given in combination with propofol for PSA [106-108]. There is no evidence that PSA using remifentanil and propofol is superior to propofol alone, nor is there evidence that either remifentanil or alfentanil is superior to fentanyl.

When used in combination with propofol for PSA, remifentanil is given in a dose of 0.5 mcg/kg (and propofol 0.5 mg/kg) over one minute [107]. Subsequent doses of remifentanil 0.25 mcg/kg and propofol 0.25 mg/kg may be given approximately every one to two minutes. When remifentanil is used ALONE for PSA, the initial dose is 0.5 to 3 mcg/kg and subsequent doses of 0.25 to 1 mcg/kg may be given approximately every two minutes as needed [106].

Few studies have assessed alfentanil as a sole agent for PSA and there are no published guidelines for its use in this manner. In one prospective observational study of 148 adults given alfentanil for PSA, 58 (39 percent) developed minor respiratory complications requiring intervention (eg, increased oxygen, brief bag-mask ventilation) despite achieving lighter levels of sedation than typically reached with propofol [109].

Alfentanil may be used as an adjunct for PSA with propofol and is given in a dose of 2.5 mcg/kg (along with propofol 0.5 mg/kg). Both may be repeated approximately every two minutes as needed.

Coadministration of midazolam and fentanyl — In settings where ultrashort-acting agents are unavailable, the combination of midazolam and fentanyl is sometimes used for PSA [34,70,110,111]. Although midazolam alone has not been shown to cause significant respiratory depression, the combination of midazolam and fentanyl can cause hypoxia and apnea, and increases the need for airway intervention and medication reversal compared with PSA using ultrashort-acting agents (eg, propofol) [34,103]. To minimize the risk of respiratory depression, we suggest that midazolam be given first and fentanyl titrated carefully thereafter, but either order is acceptable. Regardless of drug order, clinicians must titrate these medications carefully to minimize the risk of oversedation and respiratory compromise.

One reasonable approach to dosing these medications when they are used together is as follows:

Give midazolam first: 0.02 mg/kg (maximum 2 mg)

Wait two minutes and observe patient response; give second dose of midazolam if necessary

Give fentanyl: 0.5 mcg/kg

Observe patient; may repeat fentanyl dose every two minutes as necessary; titrate to effect

Use smaller doses and longer intervals between doses in the elderly and patients with compromised hepatic or renal function

Ketamine — Ketamine is a phencyclidine derivative that acts as a dissociative sedative. It produces a trance-like state and provides sedation, analgesia, and amnesia, while preserving upper airway muscle tone, airway protective reflexes, and spontaneous breathing. Because of its rapid onset, relatively short duration of action, and excellent sedative and analgesic properties, it is often used for brief, painful procedures, such as fracture reduction or laceration repair [91,112].

Ketamine is generally given IV to adults, which enables immediate onset, but it can be given intramuscularly. The duration of effect is 10 to 20 minutes. For PSA in adults, a dose of 1 to 2 mg/kg is given IV over one to two minutes. Subsequent doses of 0.25 to 1 mg/kg may be repeated every 5 to 10 minutes thereafter. If ketamine is being used as a single agent for sedation, the dose is usually at the higher end of the range, typically 0.5 to 1 mg/kg. If ketamine is being used in combination with other agents, subsequent doses are at the lower end of the range, typically 0.25 to 0.5 mg/kg. As with any sedative, the decision about appropriate repeat doses should take into consideration the effect the initial dose, the duration of the procedure, and the depth of sedation required for its completion. It is prudent to remember that more medication can always be given if needed, but excess cannot be removed.

According to a systematic review of 87 studies involving over 70,000 patients, significant adverse reactions rarely occur when ketamine is used for PSA in adults [113]. The authors emphasize that ketamine has proven to be an extremely safe drug despite being used frequently in "austere, poorly monitored settings."

The reported side effects of ketamine include tachycardia, hypertension, laryngospasm, emergence reactions, nausea and vomiting, increased intracranial and intraocular pressure, and hypersalivation [20,91,113,114]. Ketamine can exacerbate schizophrenia and should be avoided in patients with this condition [115,116]. Tachycardia and hypertension are generally mild and transient, and significant cardiorespiratory events are rare.

The risk of laryngospasm may be greater in patients with anatomic abnormalities of the upper airway (eg, tracheal stenosis, tracheomalacia) or those undergoing procedures involving significant or prolonged stimulation of the oropharynx. Guidelines published by the American College of Emergency Physicians recommend preventing secretions or blood from accumulating in the posterior oropharynx and avoiding excessive stimulation of this region with suction devices or other instruments in patients receiving ketamine for PSA [20].

Emergence reactions — Emergence reactions are the most commonly reported side effect [113]. These reactions vary in their intensity and have been described as disorientation, dream-like experiences, or hallucinations that may be frightening. They occur in up to 20 percent of adults but can be prevented or treated by giving a small dose of midazolam [3,113,117-120]. For prevention, midazolam, approximately 0.05 mg/kg (typical adult dose 2 to 4 mg), may be given slowly (over about two minutes) by IV prior to administering ketamine. A small randomized trial found that pretreatment with a fixed dose of haloperidol (5 mg) produced a comparable reduction in emergence reactions [121]. When given as pretreatments for emergence delirium, midazolam and haloperidol appear to cause no adverse clinical effects, but both slightly prolong recovery time (17 and 32 minutes, respectively) [121].

Although disturbing in some cases, emergence reactions are often benign and self-limited, requiring no pharmacologic treatment. When they occur, emergence reactions can be promptly and effectively treated with small doses of benzodiazepines such as midazolam [122,123]. Thus, whether to use pretreatment or to intervene if necessary when a reaction occurs can be left to the clinician's discretion.

Nausea and vomiting associated with ketamine administration occur in approximately 4 percent of adults. They usually occur when the patient is awake and alert and do not appear to predispose the patient to aspiration. Prophylactic treatment with midazolam has met with mixed results in pediatric populations, but pretreatment with ondansetron or comparable agents may be helpful. (See "Pharmacologic agents for pediatric procedural sedation outside of the operating room", section on 'Ketamine'.)

Ketamine can lead to hypersalivation, which can be reduced by pretreating with glycopyrrolate or atropine, although the benefit of such pretreatment is unclear [103].

Barbiturates — Barbiturates suppress the reticular activating center in the brainstem and cerebral cortex, thereby causing sedation. Methohexital is the most commonly used barbiturate for PSA but has largely been supplanted by etomidate and propofol.

Methohexital has immediate onset, a duration of action less than 10 minutes, and provides sedation and amnesia but no analgesia. It is often given in combination with opiates, which can potentiate respiratory depression. The initial dose of methohexital is 0.75 to 1 mg/kg given intravenously; repeat doses of 0.5 mg/kg IV can be given every two minutes.

Methohexital causes myocardial depression, which can lead to hypotension and tachycardia. Unlike other barbiturates, methohexital can precipitate or exacerbate seizures and should be avoided in patients with a seizure disorder [124]. One small randomized trial found methohexital and propofol to have comparable safety and efficacy when used for fracture and dislocation reduction [125].

Thiopental is a barbiturate used for induction of general anesthesia, and rarely used in the performance of PSA. It is similar in efficacy and side effects to methohexital but suppresses seizures.

Ketamine and propofol (ketofol) — "Ketofol" is a combination of ketamine and propofol being studied for use in PSA. The concept of ketofol is that the benefits of the two medications are synergistic and allow lower doses of each to be used. Lower doses purportedly reduce the risk for potential side effects (ie, propofol-induced hypotension and ketamine-induced vomiting and emergence reactions) [126,127].

According to a systematic review of six randomized trials performed in emergency departments, patients treated with a ketamine-propofol combination for procedural sedation experienced fewer adverse respiratory events compared with those treated with propofol alone (29.0 versus 35.4 percent; RR 0.82, 95% CI 0.68-0.99) [128]. However, many of these adverse events were not clinically important (eg, brief oxygen desaturation), and the review found no significant difference in the overall rate of adverse events or in the time required to complete procedures.

Another systematic review of 18 controlled trials comparing combinations of ketamine and propofol (ie, "ketofol") to ketamine or propofol alone found that ketofol may cause less respiratory depression requiring intervention and less bradycardia and hypotension than propofol alone [129]. Although the interventions required to correct respiratory problems were not described, transient respiratory depression, bradycardia, or hypotension in patients receiving PSA in the emergency department is generally not considered clinically significant. If the clinician considers it important to avoid all such events in a particular patient, the clinician should reconsider whether PSA is appropriate. (See 'Contraindications and precautions' above.)

Some of the better studies of ketofol to date have not shown this combination therapy to be more efficacious or safer than propofol or ketamine alone when performing PSA in adults [130-135]. Examples include the following:

A randomized, double-blind, multi-center trial involving 573 patients receiving PSA reported no clinically significant differences in outcome between those managed with propofol and those managed with ketofol [136]. Overall, 5 percent of patients in the propofol group and 3 percent in the ketofol group experienced the primary outcome, a respiratory adverse event (desaturation, apnea, or hypoventilation) requiring an intervention, an absolute difference of 2 percent (95% CI -2 to 5%). Patients receiving propofol were more likely to experience hypotension, although this was not clinically significant, while patients receiving ketofol were more likely to experience severe emergence delirium.

A trial performed in the emergency department (ED) of a university teaching hospital reported no significant difference in the overall incidence of respiratory depression (the primary endpoint) between patients randomly assigned to treatment with a combination of ketamine and propofol for PSA (21/97; 22 percent) or propofol alone (27/96; 28 percent) [133]. The authors report that the group given the combination of ketamine and propofol exhibited fewer episodes of apnea and oxygen desaturation and more consistent sedation, and required smaller cumulative doses of propofol. However, these differences did not achieve statistical significance and their clinical importance remains uncertain.

A randomized, double-blind trial performed in a community hospital ED, involving 284 patients requiring PSA, reported no significant difference in adverse respiratory events between patients treated with a combination of ketamine and propofol (43 events) and those given propofol alone (46 events) [137]. As with the study above, the ketofol group was reported to experience more consistent sedation, as determined by sedation scale scores and the need for additional medication, but there did not appear to be important clinical differences in outcome. Patients given ketofol had less agitation during the procedure than those given propofol (5 versus 15 patients), while the opposite was true during recovery (10 versus 7 patients).

Dexmedetomidine — Dexmedetomidine is a relatively new medication, and further research and clinical experience are needed to determine its appropriate role for procedural sedation in adults. While dexmedetomidine appears to have many favorable properties for PSA, it does not appear to have any distinct advantage over other sedative agents with regards to efficacy and side effects. On the other hand, it is routinely used for sedation of mechanically ventilated patients in intensive care units. (See "Sedative-analgesic medications in critically ill adults: Properties, dosage regimens, and adverse effects", section on 'Dexmedetomidine'.)

Marketed under brand names that include Precedex, Cepedex, and Dexdor, dexmedetomidine is an alpha agonist that acts at the locus coeruleus in the pons to reduce release of norepinephrine. This action results in sedation that is more similar to a natural sleep-like state than sedation produced by GABA-ergic agents such as propofol and benzodiazepines.

Sedation with dexmedetomidine is characterized by generally preserved muscle tone and respiratory effort (even at 10 times the maximum recommended dosage), spontaneous movement, and easy arousal, enabling the patient to obey simple instructions [138-142]. Several small studies report that decreased respiratory drive and airway obstruction may occur, albeit infrequently [143-146]. The clinician administering dexmedetomidine should remain vigilant for these uncommon but potentially serious events. Dexmedetomidine appears to reduce pain through modulation of alpha receptors in the spinal cord [147,148]. Dose-dependent bradycardia and hypotension can occur, although hypertension may be seen initially during high-dose infusion due to stimulation of alpha 2B receptors [147].

When used as a sedative agent, dexmedetomidine is usually given as an infusion at a rate of 0.2 to 0.7 mcg/kg/hour. A bolus of 0.5 to 1 mcg/kg can be given over 10 minutes prior to starting infusion. Dexmedetomidine can also be given intranasally in doses of 2 to 3 mcg/kg for anxiolysis and sedation when other routes of administration are felt not to be optimal choices [149,150].

Some small observational studies report that dexmedetomidine, when used alone, may provide unpredictable degrees of amnesia and sedation, as well as unacceptable levels of bradycardia and hypotension, often requiring termination of the procedure or use of adjunctive sedatives [151,152]. However, in a small number of trials, the combination of dexmedetomidine and other sedative agents such as ketamine and propofol provided effective procedural sedation while minimizing cardiovascular depression [153,154].

As noted elsewhere, the simultaneous administration of different classes of sedative medications (sedative-hypnotic agents, opioids, benzodiazepines) can be accomplished safely and in many instances may reduce the duration of sedation, control procedural pain, and improve amnesia. However, co-administration of multiple medications with similar mechanisms or effects can exert synergistic depressive effects on cardiovascular and respiratory function. Giving small doses titrated to effect permits safe administration of sedative medications whether they are used alone or in combination with other medications [33].

Nitrous oxide — Nitrous oxide (N2O) is an ultra-short acting agent used for PSA that is inhaled as a 30 to 50 percent mixture, with 30 percent oxygen to avoid hypoxemia. N2O has an immediate onset of action and provides analgesia, anxiolysis, and sedation. The use of N2O also obviates the need for an intravenous line. The major disadvantage to N2O is that it must be administered in a well-ventilated room with a scavenging system to prevent clinician exposure [103].

Few controlled studies about the use of N2O in adults have been published. Studies in children have generally found it to be safe, but it may not provide adequate analgesia for more painful procedures, such as fracture reduction [155].

MEDICATION SELECTION

Patients without increased risk — Procedural sedation and analgesia (PSA) is generally performed in relatively healthy patients who are hemodynamically stable. In such patients, we suggest that PSA be performed using propofol. Etomidate may also be used. The relative advantages and disadvantages of each drug are discussed immediately below (table 2). However, for well-trained clinicians experienced in the performance of PSA who are monitoring patients appropriately and prepared to deal with procedure-related complications, the best sedating medication is often the one the clinician is most comfortable using.

Propofol versus etomidate — Both propofol and etomidate have gained popularity as medications for procedural sedation and analgesia (PSA). Both medications are safe and effective in the performance of PSA, and both possess similar times to onset and recovery [90,156]. Propofol may result in a higher procedural success rate [90,130].

Factors to consider when choosing between propofol and etomidate include the following:

Etomidate provides greater hemodynamic stability, as propofol can cause hypotension. The fall in blood pressure from propofol is generally small and transient, but this difference may be of importance in patients with hypovolemia or hypotension who undergo PSA [157-159].

Etomidate can cause myoclonus, which appears to reduce the rate of procedural success [90,91,160,161].

Etomidate causes dose-dependent adrenal suppression, which may be harmful in patients with severe disease (eg, sepsis, multiple trauma). Such patients may not be suitable for PSA. This effect is unlikely to be important in otherwise healthy patients.

Respiratory depression occurs at comparable rates during PSA with both drugs, although this rarely causes harm to the patient.

Few studies have directly compared propofol and etomidate for PSA. In the largest such trial, 214 patients undergoing painful procedures in the emergency department were randomly assigned to sedation with either medication [90]. Myoclonus was more frequent with etomidate (20 versus less than 2 percent with propofol). This likely accounts for the lower rate of procedural success in the etomidate group (89 versus 97 percent). No clinically significant complications (eg, prolonged hypoxia) occurred in either group.

Propofol versus ketamine — Ketamine is more commonly used to sedate children and few studies of its role in adult procedural sedation have been performed. One small, unblinded randomized trial in adults reported higher rates of subclinical respiratory depression (as determined by changes in end-tidal carbon dioxide [EtCO2] or oxygen saturation), longer median times for return to baseline mental status (14 versus 5 minutes), and increased agitation during recovery among patients treated with ketamine for procedural sedation compared to those treated with propofol [162]. The rates for clinical intervention for respiratory problems, duration of the procedure, and successful completion of the procedure did not differ between the two groups.

Ketamine plus propofol ("ketofol") — While available evidence suggests that ketofol is a safe and effective drug combination for procedural sedation, there is no convincing evidence that it improves clinically significant outcomes or reduces important complications during PSA (which are rare) compared to propofol.

Patients at increased risk — In some circumstances, clinicians, after carefully considering the relative risks and benefits, may elect to perform PSA in patients at increased risk of complications. Suggestions for drug selection in several common scenarios are provided here (table 2). Contraindications to PSA and considerations when performing PSA during pregnancy and in obese patients are discussed in greater detail above. (See 'Contraindications and precautions' above and 'Considerations in pregnancy' above and 'Considerations in obese adults' above.)

Patients at risk of hypotension — In patients at risk of hypotension due to recent illness and dehydration, cardiac disease, or some other condition, we suggest that either etomidate or ketamine be used for PSA; in contrast, propofol has a greater blood pressure lowering effect [90]. Either agent will maintain hemodynamic stability. (See 'Etomidate' above and 'Ketamine' above.)

Patient at risk for airway or respiratory complications — In patients who may have a potentially difficult airway to manage or have compromised respiratory function, we suggest that ketamine be used for PSA. Ketamine allows the patient to maintain protective airway reflexes and does not cause respiratory depression.

Older adult patients — Older adult patients are at increased risk of complications during PSA [10]. As a result, sedatives administered to older patients for PSA, regardless of the agent, should be given at a lower starting dose with slower rates of administration and less frequent dosing intervals. In older adult patients without major comorbidities or hemodynamic instability, it may be best to perform PSA using an ultrashort-acting sedative, such as propofol. Procedures in older adult patients with major comorbidities are probably best performed in the operating room. (See 'Propofol' above.)

Ketamine versus short-acting opioids for analgesia — Ultra short-acting opioids (eg, fentanyl) provide analgesia during PSA but can contribute to respiratory depression. Some researchers hypothesize that a sub-dissociative dose of ketamine can provide adequate analgesia without the risk of respiratory depression.

One small randomized trial found that patients given ketamine (0.3 mg/kg IV) and propofol during PSA achieved similar analgesia but experienced fewer complications compared with patients given fentanyl (1.5 mcg/kg IV) and propofol, who had five times the risk of experiencing a serious adverse event (eg, hypoxia) (95% CI 1.9-13.6) [127]. A smaller, nonrandomized trial found that the addition of ketamine reduced pain and the need for post-procedural analgesics [163]. Further studies are needed to confirm these preliminary findings.

DISCHARGE CRITERIA — There is little evidence to guide decisions about discharge following procedural sedation and analgesia (PSA). Some guidelines suggest that patients are ready for discharge when they have reached their "neuromuscular and cognitive pre-procedure baseline" [164].

Certain conditions should be met before a patient can be considered safe for discharge following PSA:

The procedure should be of sufficiently low risk that additional monitoring for complications is unnecessary.

Symptoms, such as pain, lightheadedness, and nausea should be well-controlled.

Vital signs and respiratory and cardiac function should be stable.

Mental status and physical function should have returned to a point where the patient can care for himself or herself with minimal to no assistance.

A reliable person who can provide support and supervision should be present at the patient's home for at least a few hours.

Clear written discharge instructions should be given and explained to the patient and to the family member or friend who will be assisting with the patient's care following PSA. The clinician should explain what was done, the expected course, potential problems, what to do if problems arise, when and where to follow up, and when to return to normal activities.

One important issue is to determine a period of observation after which adverse events are sufficiently unlikely that even a patient who has not entirely returned to baseline (eg, exhibits mild drowsiness) can be discharged safely. The only study to examine the timing of significant adverse events associated with PSA was performed in a sample of 1341 children and found that 92 percent of such events occurred during the procedure, while only 8 percent occurred afterwards [164]. Significant adverse events rarely occurred more than 25 minutes after the last dose of medication was administered, provided that no adverse events had occurred up to that point. Rarely, episodes of hypoxia developed up to 40 minutes after the last medication dose. In all such cases, prior episodes of hypoxia had occurred.

Based on these data, some clinicians believe that patients can be safely discharged within 30 minutes of receiving their last dose of sedative provided that no significant adverse events occurred during the procedure and the patient did not receive a reversal agent (eg, naloxone). Of note, only relatively long acting agents, such as ketamine, midazolam and fentanyl were used in this study (ie, neither etomidate nor propofol were used). It is unlikely that a longer period of observation would be necessary following PSA with ultra-short acting agents [13].

Although serious adverse events, such as hypoxia, rarely occur after discharge, it is not uncommon for patients to experience mild symptoms, such as nausea, lightheadedness, fatigue, or unsteadiness, for up to 24 hours [6,165,166]. This should be made clear to the patient.

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Procedural sedation in adults".)

SUMMARY AND RECOMMENDATIONS

Procedural sedation and analgesia (PSA) involves the use of short-acting analgesic and sedative medications to enable clinicians to perform procedures, while monitoring the patient closely for potential adverse effects. This process was previously (and inappropriately) termed "conscious sedation." (See 'Definitions' above.)

PSA may be used for any procedure in which a patient's pain or anxiety may be excessive and may impede performance. There are no absolute contraindications to PSA. Relative contraindications include: older age, significant medical comorbidities, and signs of a difficult airway. Whether the patient recently ate should be considered before performing PSA, although this may not increase aspiration risk. (See 'Indications' above and 'Contraindications and precautions' above.)

The number of clinicians needed to perform PSA and the procedure safely may vary according to the patient and the procedure. In most cases, one clinician performs the procedure while another (usually a nurse) administers the sedative agents and monitors and records the patient's vital signs and clinical status. Whenever possible, we suggest that this minimum standard be met. (See 'Prerequisites and personnel' above.)

Proper monitoring during PSA is crucial. The patient's blood pressure, heart rate, and respiratory rate should be measured at frequent, regular intervals; oxygen saturation (SpO2), end-tidal carbon dioxide (EtCO2) level, and cardiac rhythm should be monitored continuously. We suggest that high flow oxygen (15 L by face mask) be provided to patients receiving PSA. (See 'Monitoring and preoxygenation' above.)

Serious complications attributable to PSA rarely occur. Significant respiratory compromise develops in less than 1 percent of cases. Adverse outcomes may include respiratory depression with hypoxia or hypercarbia, cardiovascular instability, vomiting and aspiration, and inadequate sedation preventing completion of the procedure. All equipment and medications necessary for airway management should be at the bedside during PSA. (See 'Complications' above and 'Equipment' above.)

Ideal drugs for PSA have a rapid onset and short duration of action, maintain hemodynamic stability, and do not cause major side effects. Several medications are commonly used and no single drug is ideal for all situations (table 2). Medications used for PSA are discussed in the text. (See 'Medications' above.)

PSA is most often performed in patients without major comorbidities or hemodynamic instability. In such patients, we suggest that PSA be performed using propofol (Grade 2B). Etomidate may also be used. The relative advantages and disadvantages of each drug are discussed above. (See 'Patients without increased risk' above.)

Older patients are at increased risk of complications during PSA. Therefore, sedatives administered to older patients for PSA, regardless of the agent, should be given using a lower starting dose, and with slower rates of administration and less frequent dosing intervals. (See 'Older adult patients' above.)

In some circumstances, clinicians, after carefully considering the relative risks and benefits, may elect to perform PSA in patients at some increased risk of complications. In patients at risk of hypotension, we suggest that either etomidate or ketamine be used for PSA (Grade 2C). In patients who may have a potentially difficult airway or have compromised respiratory function, we suggest that ketamine be used for PSA (Grade 2C). (See 'Patients at increased risk' above.)

Criteria for safe discharge following PSA are described in the text. (See 'Discharge criteria' above.)

REFERENCES

  1. Green SM, Krauss B. Procedural sedation terminology: moving beyond "conscious sedation". Ann Emerg Med 2002; 39:433.
  2. Green SM, Irwin MG, Mason KP, International Committee for the Advancement of Procedural Sedation. Procedural sedation: providing the missing definition. Anaesthesia 2021; 76:598.
  3. Gan TJ. Pharmacokinetic and pharmacodynamic characteristics of medications used for moderate sedation. Clin Pharmacokinet 2006; 45:855.
  4. American Society of Anesthesiologists Task Force on Sedation and Analgesia by Non-Anesthesiologists. Practice guidelines for sedation and analgesia by non-anesthesiologists. Anesthesiology 2002; 96:1004.
  5. Joint Commission on Accreditation of Healthcare Organizations. Comprehensive Accreditation Manual for Hospitals, The Official Handbood. JCAHO Publication, Chicago 2004.
  6. Krauss B, Green SM. Sedation and analgesia for procedures in children. N Engl J Med 2000; 342:938.
  7. Brady M, Kinn S, Stuart P. Preoperative fasting for adults to prevent perioperative complications. Cochrane Database Syst Rev 2003; :CD004423.
  8. Cheung KW, Watson ML, Field S, Campbell SG. Aspiration pneumonitis requiring intubation after procedural sedation and analgesia: a case report. Ann Emerg Med 2007; 49:462.
  9. Sakai T, Planinsic RM, Quinlan JJ, et al. The incidence and outcome of perioperative pulmonary aspiration in a university hospital: a 4-year retrospective analysis. Anesth Analg 2006; 103:941.
  10. Mace SE. Adverse events of emergency department procedural sedation. [abstract] Ann Emerg Med 2006; 48:S59.
  11. Miller MA, Levy P, Patel MM. Procedural sedation and analgesia in the emergency department: what are the risks? Emerg Med Clin North Am 2005; 23:551.
  12. Cepeda MS, Farrar JT, Baumgarten M, et al. Side effects of opioids during short-term administration: effect of age, gender, and race. Clin Pharmacol Ther 2003; 74:102.
  13. Miner JR, Burton JH. Clinical practice advisory: Emergency department procedural sedation with propofol. Ann Emerg Med 2007; 50:182.
  14. Green SM, Mason KP, Krauss BS. Pulmonary aspiration during procedural sedation: a comprehensive systematic review. Br J Anaesth 2017; 118:344.
  15. Godwin SA, Burton JH, Gerardo CJ, et al. Clinical policy: procedural sedation and analgesia in the emergency department. Ann Emerg Med 2014; 63:247.
  16. Green SM, Leroy PL, Roback MG, et al. An international multidisciplinary consensus statement on fasting before procedural sedation in adults and children. Anaesthesia 2020; 75:374.
  17. Green SM, Roback MG, Krauss BS, et al. Unscheduled Procedural Sedation: A Multidisciplinary Consensus Practice Guideline. Ann Emerg Med 2019; 73:e51.
  18. Bassett KE, Anderson JL, Pribble CG, Guenther E. Propofol for procedural sedation in children in the emergency department. Ann Emerg Med 2003; 42:773.
  19. Green SM, Roback MG, Miner JR, et al. Fasting and emergency department procedural sedation and analgesia: a consensus-based clinical practice advisory. Ann Emerg Med 2007; 49:454.
  20. Green SM, Roback MG, Kennedy RM, Krauss B. Clinical practice guideline for emergency department ketamine dissociative sedation: 2011 update. Ann Emerg Med 2011; 57:449.
  21. Coté CJ, Wilson S, AMERICAN ACADEMY OF PEDIATRICS, AMERICAN ACADEMY OF PEDIATRIC DENTISTRY. Guidelines for Monitoring and Management of Pediatric Patients Before, During, and After Sedation for Diagnostic and Therapeutic Procedures: Update 2016. Pediatrics 2016; 138.
  22. Sandhar BK, Goresky GV, Maltby JR, Shaffer EA. Effect of oral liquids and ranitidine on gastric fluid volume and pH in children undergoing outpatient surgery. Anesthesiology 1989; 71:327.
  23. Brady M, Kinn S, O'Rourke K, et al. Preoperative fasting for preventing perioperative complications in children. Cochrane Database Syst Rev 2005; :CD005285.
  24. Engelhardt T, Webster NR. Pulmonary aspiration of gastric contents in anaesthesia. Br J Anaesth 1999; 83:453.
  25. Green SM, Krauss B. Pulmonary aspiration risk during emergency department procedural sedation--an examination of the role of fasting and sedation depth. Acad Emerg Med 2002; 9:35.
  26. Søreide E, Eriksson LI, Hirlekar G, et al. Pre-operative fasting guidelines: an update. Acta Anaesthesiol Scand 2005; 49:1041.
  27. Ingebo KR, Rayhorn NJ, Hecht RM, et al. Sedation in children: adequacy of two-hour fasting. J Pediatr 1997; 131:155.
  28. Bell A, Treston G, McNabb C, et al. Profiling adverse respiratory events and vomiting when using propofol for emergency department procedural sedation. Emerg Med Australas 2007; 19:405.
  29. Ghaffar S, Haverland C, Ramaciotti C, et al. Sedation for pediatric echocardiography: evaluation of preprocedure fasting guidelines. J Am Soc Echocardiogr 2002; 15:980.
  30. Splinter WM, Stewart JA, Muir JG. Large volumes of apple juice preoperatively do not affect gastric pH and volume in children. Can J Anaesth 1990; 37:36.
  31. Olsson GL, Hallen B, Hambraeus-Jonzon K. Aspiration during anaesthesia: a computer-aided study of 185,358 anaesthetics. Acta Anaesthesiol Scand 1986; 30:84.
  32. Blitt CD, Gutman HL, Cohen DD, et al. "Silent" regurgitation and aspiration during general anesthesia. Anesth Analg 1970; 49:707.
  33. Practice Guidelines for Moderate Procedural Sedation and Analgesia 2018: A Report by the American Society of Anesthesiologists Task Force on Moderate Procedural Sedation and Analgesia, the American Association of Oral and Maxillofacial Surgeons, American College of Radiology, American Dental Association, American Society of Dentist Anesthesiologists, and Society of Interventional Radiology. Anesthesiology 2018; 128:437.
  34. Godwin SA, Caro DA, Wolf SJ, et al. Clinical policy: procedural sedation and analgesia in the emergency department. Ann Emerg Med 2005; 45:177.
  35. Harrington L. Nurse-administered propofol sedation: a review of current evidence. Gastroenterol Nurs 2006; 29:371.
  36. Coté CJ, Notterman DA, Karl HW, et al. Adverse sedation events in pediatrics: a critical incident analysis of contributing factors. Pediatrics 2000; 105:805.
  37. Hogan K, Sacchetti A, Aman L, Opiela D. The safety of single-physician procedural sedation in the emergency department. Emerg Med J 2006; 23:922.
  38. Deitch K, Chudnofsky CR, Dominici P, et al. The utility of high-flow oxygen during emergency department procedural sedation and analgesia with propofol: a randomized, controlled trial. Ann Emerg Med 2011; 58:360.
  39. Deitch K, Chudnofsky CR, Dominici P. The utility of supplemental oxygen during emergency department procedural sedation and analgesia with midazolam and fentanyl: a randomized, controlled trial. Ann Emerg Med 2007; 49:1.
  40. Miner JR, Biros MH, Heegaard W, Plummer D. Bispectral electroencephalographic analysis of patients undergoing procedural sedation in the emergency department. Acad Emerg Med 2003; 10:638.
  41. Miner JR, Heegaard W, Plummer D. End-tidal carbon dioxide monitoring during procedural sedation. Acad Emerg Med 2002; 9:275.
  42. McQuillen KK, Steele DW. Capnography during sedation/analgesia in the pediatric emergency department. Pediatr Emerg Care 2000; 16:401.
  43. Rozario L, Sloper D, Sheridan MJ. Supplemental oxygen during moderate sedation and the occurrence of clinically significant desaturation during endoscopic procedures. Gastroenterol Nurs 2008; 31:281.
  44. Sivilotti ML, Messenger DW, van Vlymen J, et al. A comparative evaluation of capnometry versus pulse oximetry during procedural sedation and analgesia on room air. CJEM 2010; 12:397.
  45. Wall BF, Magee K, Campbell SG, Zed PJ. Capnography versus standard monitoring for emergency department procedural sedation and analgesia. Cochrane Database Syst Rev 2017; 3:CD010698.
  46. Deitch K, Chudnofsky CR, Dominici P. Can emergency physicians recognize respiratory depression in patients who do not become hypoxic during ED procedural sedation and analgesia. Ann Emerg Med 2007; 50:S122.
  47. Deitch K, Miner J, Chudnofsky CR, et al. Does end tidal CO2 monitoring during emergency department procedural sedation and analgesia with propofol decrease the incidence of hypoxic events? A randomized, controlled trial. Ann Emerg Med 2010; 55:258.
  48. Wright SW. Conscious sedation in the emergency department: the value of capnography and pulse oximetry. Ann Emerg Med 1992; 21:551.
  49. Gill M, Green SM, Krauss B. A study of the Bispectral Index Monitor during procedural sedation and analgesia in the emergency department. Ann Emerg Med 2003; 41:234.
  50. American Society of Anesthesiologists Task Force on Intraoperative Awareness. Practice advisory for intraoperative awareness and brain function monitoring: a report by the american society of anesthesiologists task force on intraoperative awareness. Anesthesiology 2006; 104:847.
  51. Niedhart DJ, Kaiser HA, Jacobsohn E, et al. Intrapatient reproducibility of the BISxp monitor. Anesthesiology 2006; 104:242.
  52. Jirapinyo P, Thompson CC. Sedation Challenges: Obesity and Sleep Apnea. Gastrointest Endosc Clin N Am 2016; 26:527.
  53. Correa J, Ornaque I, Martí-Valeri C. Predictors of difficult airway management in morbid obesity patients. Eur J Anaesthesiol 2006; 23:263.
  54. Shailaja S, Nichelle SM, Shetty AK, Hegde BR. Comparing ease of intubation in obese and lean patients using intubation difficulty scale. Anesth Essays Res 2014; 8:168.
  55. Wani S, Azar R, Hovis CE, et al. Obesity as a risk factor for sedation-related complications during propofol-mediated sedation for advanced endoscopic procedures. Gastrointest Endosc 2011; 74:1238.
  56. Bautista A, Liu R, OngSio LC, Wadhwa A. Pharmacotherapy of procedural sedation in morbidly obese. J Clin Res Pain Anesth 2020; 2:180018.
  57. Cortínez LI, Anderson BJ, Penna A, et al. Influence of obesity on propofol pharmacokinetics: derivation of a pharmacokinetic model. Br J Anaesth 2010; 105:448.
  58. Erstad BL, Barletta JF. Drug dosing in the critically ill obese patient-a focus on sedation, analgesia, and delirium. Crit Care 2020; 24:315.
  59. Ingrande J, Lemmens HJ. Anesthetic Pharmacology and the Morbidly Obese Patient. Curr Anesthesiol Rep 2013; 3:10.
  60. Tu W, Zhou D, Li Z, et al. Dexmedetomidine pharmacokinetics in morbidly obese patients. American Society of Anesthesiologists (ASA) 2015. http://www.asaabstracts.com/strands/asaabstracts/abstract.htm?year=2015&index=7&absnum=4447.
  61. De Baerdemaeker L, Mortier E, Struys M. Pharmacokinetics in obese patients. Contin Educ Anaesth Crit Care Pain 2004; 93:645.
  62. Bellolio MF, Gilani WI, Barrionuevo P, et al. Incidence of Adverse Events in Adults Undergoing Procedural Sedation in the Emergency Department: A Systematic Review and Meta-analysis. Acad Emerg Med 2016; 23:119.
  63. Miller KA, Andolfatto G, Miner JR, et al. Clinical Practice Guideline for Emergency Department Procedural Sedation With Propofol: 2018 Update. Ann Emerg Med 2019; 73:470.
  64. Peña BM, Krauss B. Adverse events of procedural sedation and analgesia in a pediatric emergency department. Ann Emerg Med 1999; 34:483.
  65. Rothermel LK. Newer pharmacologic agents for procedural sedation of children in the emergency department-etomidate and propofol. Curr Opin Pediatr 2003; 15:200.
  66. Skokan EG, Pribble C, Bassett KE, Nelson DS. Use of propofol sedation in a pediatric emergency department: a prospective study. Clin Pediatr (Phila) 2001; 40:663.
  67. Clinical policy for procedural sedation and analgesia in the emergency department. American College of Emergency Physicians. Ann Emerg Med 1998; 31:663.
  68. Tomicheck RC, Rosow CE, Philbin DM, et al. Diazepam-fentanyl interaction--hemodynamic and hormonal effects in coronary artery surgery. Anesth Analg 1983; 62:881.
  69. Blackburn P, Vissers R. Pharmacology of emergency department pain management and conscious sedation. Emerg Med Clin North Am 2000; 18:803.
  70. Pitetti RD, Singh S, Pierce MC. Safe and efficacious use of procedural sedation and analgesia by nonanesthesiologists in a pediatric emergency department. Arch Pediatr Adolesc Med 2003; 157:1090.
  71. Vinson DR, Bradbury DR. Etomidate for procedural sedation in emergency medicine. Ann Emerg Med 2002; 39:592.
  72. Chew ST, Ip-Yam PC, Kong CF. Recovery following tonsillectomy a comparison between tramadol and morphine for analgesia. Singapore Med J 2003; 44:296.
  73. Roback MG, Wathen JE, Bajaj L, Bothner JP. Adverse events associated with procedural sedation and analgesia in a pediatric emergency department: a comparison of common parenteral drugs. Acad Emerg Med 2005; 12:508.
  74. Langston WT, Wathen JE, Roback MG, Bajaj L. Effect of ondansetron on the incidence of vomiting associated with ketamine sedation in children: a double-blind, randomized, placebo-controlled trial. Ann Emerg Med 2008; 52:30.
  75. Swanson ER, Seaberg DC, Mathias S. The use of propofol for sedation in the emergency department. Acad Emerg Med 1996; 3:234.
  76. Diprivan (propofol): manufacturer's package insert. US National Library of Medicine www.dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?id=4756#section-11 (Accessed on May 26, 2009).
  77. Jalota L, Kalira V, George E, et al. Prevention of pain on injection of propofol: systematic review and meta-analysis. BMJ 2011; 342:d1110.
  78. Koo SW, Cho SJ, Kim YK, et al. Small-dose ketamine reduces the pain of propofol injection. Anesth Analg 2006; 103:1444.
  79. Miner JR, Gray RO, Stephens D, Biros MH. Randomized clinical trial of propofol with and without alfentanil for deep procedural sedation in the emergency department. Acad Emerg Med 2009; 16:825.
  80. Optimizing the Treatment of Acute Pain in the Emergency Department. Ann Emerg Med 2017; 70:446.
  81. Kurdi MS, Theerth KA, Deva RS. Ketamine: Current applications in anesthesia, pain, and critical care. Anesth Essays Res 2014; 8:283.
  82. Newstead B, Bradburn S, Appelboam A, et al. Propofol for adult procedural sedation in a UK emergency department: safety profile in 1008 cases. Br J Anaesth 2013; 111:651.
  83. Black E, Campbell SG, Magee K, Zed PJ. Propofol for procedural sedation in the emergency department: a qualitative systematic review. Ann Pharmacother 2013; 47:856.
  84. Walker JA, McIntyre RD, Schleinitz PF, et al. Nurse-administered propofol sedation without anesthesia specialists in 9152 endoscopic cases in an ambulatory surgery center. Am J Gastroenterol 2003; 98:1744.
  85. Tohda G, Higashi S, Sakumoto H, et al. Efficacy and safety of nurse-administered propofol sedation during emergency upper endoscopy for gastrointestinal bleeding: a prospective study. Endoscopy 2006; 38:684.
  86. Wakai A, Blackburn C, McCabe A, et al. The use of propofol for procedural sedation in emergency departments. Cochrane Database Syst Rev 2015; :CD007399.
  87. Green SM, Krauss B. Barriers to propofol use in emergency medicine. Ann Emerg Med 2008; 52:392.
  88. Flynn G. "Poachers and dabblers?": ASA president's incautious comment riles emergency physicians. Ann Emerg Med 2007; 50:264.
  89. Weaver JM. The great debate on nurse-administered propofol sedation (NAPS)--where should we stand? Anesth Prog 2006; 53:31.
  90. Miner JR, Danahy M, Moch A, Biros M. Randomized clinical trial of etomidate versus propofol for procedural sedation in the emergency department. Ann Emerg Med 2007; 49:15.
  91. Brown TB, Lovato LM, Parker D. Procedural sedation in the acute care setting. Am Fam Physician 2005; 71:85.
  92. Falk J, Zed PJ. Etomidate for procedural sedation in the emergency department. Ann Pharmacother 2004; 38:1272.
  93. Sacchetti A, Senula G, Strickland J, Dubin R. Procedural sedation in the community emergency department: initial results of the ProSCED registry. Acad Emerg Med 2007; 14:41.
  94. Keim SM, Erstad BL, Sakles JC, Davis V. Etomidate for procedural sedation in the emergency department. Pharmacotherapy 2002; 22:586.
  95. Doenicke AW, Roizen MF, Kugler J, et al. Reducing myoclonus after etomidate. Anesthesiology 1999; 90:113.
  96. Burton JH, Bock AJ, Strout TD, Marcolini EG. Etomidate and midazolam for reduction of anterior shoulder dislocation: a randomized, controlled trial. Ann Emerg Med 2002; 40:496.
  97. Ruth WJ, Burton JH, Bock AJ. Intravenous etomidate for procedural sedation in emergency department patients. Acad Emerg Med 2001; 8:13.
  98. Hüter L, Schreiber T, Gugel M, Schwarzkopf K. Low-dose intravenous midazolam reduces etomidate-induced myoclonus: a prospective, randomized study in patients undergoing elective cardioversion. Anesth Analg 2007; 105:1298.
  99. Guler A, Satilmis T, Akinci SB, et al. Magnesium sulfate pretreatment reduces myoclonus after etomidate. Anesth Analg 2005; 101:705.
  100. Absalom A, Pledger D, Kong A. Adrenocortical function in critically ill patients 24 h after a single dose of etomidate. Anaesthesia 1999; 54:861.
  101. Schenarts CL, Burton JH, Riker RR. Adrenocortical dysfunction following etomidate induction in emergency department patients. Acad Emerg Med 2001; 8:1.
  102. Horn E, Nesbit SA. Pharmacology and pharmacokinetics of sedatives and analgesics. Gastrointest Endosc Clin N Am 2004; 14:247.
  103. Bahn EL, Holt KR. Procedural sedation and analgesia: a review and new concepts. Emerg Med Clin North Am 2005; 23:503.
  104. Muse DA. Conscious and deep sedation. In: The Clinical Practice of Emergency Medicine, 3rd ed, Harwood-Nuss, A, Wolfson, AB (Eds), Lippincott, Williams and Wilkins, Philadelphia 2001. p.1761.
  105. Kisilewicz M, Rosenberg H, Vaillancourt C. Remifentanil for procedural sedation: a systematic review of the literature. Emerg Med J 2017; 34:294.
  106. Phillips WJ, Halpin J, Jones J, McKenzie K. Remifentanil for procedural sedation in the emergency department. Ann Emerg Med 2009; 53:163.
  107. Dunn MJ, Mitchell R, Souza CD, Drummond G. Evaluation of propofol and remifentanil for intravenous sedation for reducing shoulder dislocations in the emergency department. Emerg Med J 2006; 23:57.
  108. Krauss B, Green SM. Procedural sedation and analgesia in children. Lancet 2006; 367:766.
  109. Miner JR, Gray R, Delavari P, et al. Alfentanil for procedural sedation in the emergency department. Ann Emerg Med 2011; 57:117.
  110. Campbell SG, Magee KD, Kovacs GJ, et al. Procedural sedation and analgesia in a Canadian adult tertiary care emergency department: a case series. CJEM 2006; 8:85.
  111. Ab-Rahman NH, Hashim A. A randomized controlled trial on procedural sedation among adult patients in emergency departments: comparing fentanyl with midazolam versus fentanyl with propofol [abstract]. Ann Emerg Med 2008; 51:479.
  112. Johnston MN, Liebelt EL. Acute pain management and procedural sedation in children. In: Emergency Medicine: A Comprehensive Study Guide, 6th Ed, Tintinalli, JE, Kelen, GD, Stapczynski, JS (Eds), McGraw-Hill, New York 2004. p.858.
  113. Strayer RJ, Nelson LS. Adverse events associated with ketamine for procedural sedation in adults. Am J Emerg Med 2008; 26:985.
  114. Newton A, Fitton L. Intravenous ketamine for adult procedural sedation in the emergency department: a prospective cohort study. Emerg Med J 2008; 25:498.
  115. Lahti AC, Koffel B, LaPorte D, Tamminga CA. Subanesthetic doses of ketamine stimulate psychosis in schizophrenia. Neuropsychopharmacology 1995; 13:9.
  116. Malhotra AK, Pinals DA, Adler CM, et al. Ketamine-induced exacerbation of psychotic symptoms and cognitive impairment in neuroleptic-free schizophrenics. Neuropsychopharmacology 1997; 17:141.
  117. Mason KP, Michna E, DiNardo JA, et al. Evolution of a protocol for ketamine-induced sedation as an alternative to general anesthesia for interventional radiologic procedures in pediatric patients. Radiology 2002; 225:457.
  118. Chudnofsky CR, Weber JE, Stoyanoff PJ, et al. A combination of midazolam and ketamine for procedural sedation and analgesia in adult emergency department patients. Acad Emerg Med 2000; 7:228.
  119. Vardy JM, Dignon N, Mukherjee N, et al. Audit of the safety and effectiveness of ketamine for procedural sedation in the emergency department. Emerg Med J 2008; 25:579.
  120. Sener S, Eken C, Schultz CH, et al. Ketamine with and without midazolam for emergency department sedation in adults: a randomized controlled trial. Ann Emerg Med 2011; 57:109.
  121. Akhlaghi N, Payandemehr P, Yaseri M, et al. Premedication With Midazolam or Haloperidol to Prevent Recovery Agitation in Adults Undergoing Procedural Sedation With Ketamine: A Randomized Double-Blind Clinical Trial. Ann Emerg Med 2019; 73:462.
  122. Green SM, Kuppermann N, Rothrock SG, et al. Predictors of adverse events with intramuscular ketamine sedation in children. Ann Emerg Med 2000; 35:35.
  123. Green SM, Clem KJ, Rothrock SG. Ketamine safety profile in the developing world: survey of practitioners. Acad Emerg Med 1996; 3:598.
  124. Nicolaou DD. Procedural sedation and analgesia. In: Emergency Medicine: A Comprehensive Study Guide, 6th Ed, Tintinalli, JE, Kelen, GK, Stapczynski, JS (Eds), McGraw-Hill, New York 2004. p.275.
  125. Miner JR, Biros M, Krieg S, et al. Randomized clinical trial of propofol versus methohexital for procedural sedation during fracture and dislocation reduction in the emergency department. Acad Emerg Med 2003; 10:931.
  126. Willman EV, Andolfatto G. A prospective evaluation of "ketofol" (ketamine/propofol combination) for procedural sedation and analgesia in the emergency department. Ann Emerg Med 2007; 49:23.
  127. Messenger DW, Murray HE, Dungey PE, et al. Subdissociative-dose ketamine versus fentanyl for analgesia during propofol procedural sedation: a randomized clinical trial. Acad Emerg Med 2008; 15:877.
  128. Yan JW, McLeod SL, Iansavitchene A. Ketamine-Propofol Versus Propofol Alone for Procedural Sedation in the Emergency Department: A Systematic Review and Meta-analysis. Acad Emerg Med 2015; 22:1003.
  129. Jalili M, Bahreini M, Doosti-Irani A, et al. Ketamine-propofol combination (ketofol) vs propofol for procedural sedation and analgesia: systematic review and meta-analysis. Am J Emerg Med 2016; 34:558.
  130. Green SM. Research advances in procedural sedation and analgesia. Ann Emerg Med 2007; 49:31.
  131. Loh G, Dalen D. Low-dose ketamine in addition to propofol for procedural sedation and analgesia in the emergency department. Ann Pharmacother 2007; 41:485.
  132. Aurora, S. Combining ketamine and propofol ("ketofol") for emergency department procedural sedation and analgesia: a review. West J Emerg Med 2008; 9: Article 5 repositories.cdlib.org/uciem/westjem/vol9/iss1/art5 (Accessed on June 17, 2009).
  133. David H, Shipp J. A randomized controlled trial of ketamine/propofol versus propofol alone for emergency department procedural sedation. Ann Emerg Med 2011; 57:435.
  134. Phillips W, Anderson A, Rosengreen M, et al. Propofol versus propofol/ketamine for brief painful procedures in the emergency department: clinical and bispectral index scale comparison. J Pain Palliat Care Pharmacother 2010; 24:349.
  135. Miner JR, Moore JC, Austad EJ, et al. Randomized, double-blinded, clinical trial of propofol, 1:1 propofol/ketamine, and 4:1 propofol/ketamine for deep procedural sedation in the emergency department. Ann Emerg Med 2015; 65:479.
  136. Ferguson I, Bell A, Treston G, et al. Propofol or Ketofol for Procedural Sedation and Analgesia in Emergency Medicine-The POKER Study: A Randomized Double-Blind Clinical Trial. Ann Emerg Med 2016; 68:574.
  137. Andolfatto G, Abu-Laban RB, Zed PJ, et al. Ketamine-propofol combination (ketofol) versus propofol alone for emergency department procedural sedation and analgesia: a randomized double-blind trial. Ann Emerg Med 2012; 59:504.
  138. Kaur M, Singh PM. Current role of dexmedetomidine in clinical anesthesia and intensive care. Anesth Essays Res 2011; 5:128.
  139. Venn RM, Hell J, Grounds RM. Respiratory effects of dexmedetomidine in the surgical patient requiring intensive care. Crit Care 2000; 4:302.
  140. Hsu YW, Cortinez LI, Robertson KM, et al. Dexmedetomidine pharmacodynamics: part I: crossover comparison of the respiratory effects of dexmedetomidine and remifentanil in healthy volunteers. Anesthesiology 2004; 101:1066.
  141. Hall JE, Uhrich TD, Barney JA, et al. Sedative, amnestic, and analgesic properties of small-dose dexmedetomidine infusions. Anesth Analg 2000; 90:699.
  142. Ebert TJ, Hall JE, Barney JA, et al. The effects of increasing plasma concentrations of dexmedetomidine in humans. Anesthesiology 2000; 93:382.
  143. Lodenius Å, Maddison KJ, Lawther BK, et al. Upper Airway Collapsibility during Dexmedetomidine and Propofol Sedation in Healthy Volunteers: A Nonblinded Randomized Crossover Study. Anesthesiology 2019; 131:962.
  144. Mahmoud M, Jung D, Salisbury S, et al. Effect of increasing depth of dexmedetomidine and propofol anesthesia on upper airway morphology in children and adolescents with obstructive sleep apnea. J Clin Anesth 2013; 25:529.
  145. Mahmoud M, Gunter J, Donnelly LF, et al. A comparison of dexmedetomidine with propofol for magnetic resonance imaging sleep studies in children. Anesth Analg 2009; 109:745.
  146. Lodenius Å, Ebberyd A, Hårdemark Cedborg A, et al. Sedation with Dexmedetomidine or Propofol Impairs Hypoxic Control of Breathing in Healthy Male Volunteers: A Nonblinded, Randomized Crossover Study. Anesthesiology 2016; 125:700.
  147. Scott-Warren VL, Sebastian J. Dexmedetomodine: its use in intensive care medicine and anesthesia. BJA Education 2015; 16:242.
  148. Arcangeli A, D'Alò C, Gaspari R. Dexmedetomidine use in general anaesthesia. Curr Drug Targets 2009; 10:687.
  149. Demiraran Y, Korkut E, Tamer A, et al. The comparison of dexmedetomidine and midazolam used for sedation of patients during upper endoscopy: A prospective, randomized study. Can J Gastroenterol 2007; 21:25.
  150. Jense RJ, Souter K, Davies J, et al. Dexmedetomidine sedation for laryngeal framework surgery. Ann Otol Rhinol Laryngol 2008; 117:659.
  151. Munro HM, Tirotta CF, Felix DE, et al. Initial experience with dexmedetomidine for diagnostic and interventional cardiac catheterization in children. Paediatr Anaesth 2007; 17:109.
  152. Jalowiecki P, Rudner R, Gonciarz M, et al. Sole use of dexmedetomidine has limited utility for conscious sedation during outpatient colonoscopy. Anesthesiology 2005; 103:269.
  153. Tosun Z, Akin A, Guler G, et al. Dexmedetomidine-ketamine and propofol-ketamine combinations for anesthesia in spontaneously breathing pediatric patients undergoing cardiac catheterization. J Cardiothorac Vasc Anesth 2006; 20:515.
  154. Tobias JD. Dexmedetomidine and ketamine: an effective alternative for procedural sedation? Pediatr Crit Care Med 2012; 13:423.
  155. Babl FE, Oakley E, Puspitadewi A, Sharwood LN. Limited analgesic efficacy of nitrous oxide for painful procedures in children. Emerg Med J 2008; 25:717.
  156. Gillies GW, Lees NW. The effects of speed of injection on induction with propofol. A comparison with etomidate. Anaesthesia 1989; 44:386.
  157. Burton JH, Miner JR, Shipley ER, et al. Propofol for emergency department procedural sedation and analgesia: a tale of three centers. Acad Emerg Med 2006; 13:24.
  158. Guenther E, Pribble CG, Junkins EP Jr, et al. Propofol sedation by emergency physicians for elective pediatric outpatient procedures. Ann Emerg Med 2003; 42:783.
  159. Coll-Vinent B, Sala X, Fernández C, et al. Sedation for cardioversion in the emergency department: analysis of effectiveness in four protocols. Ann Emerg Med 2003; 42:767.
  160. Plewa MC, King R, Johnson D, et al. Etomidate use during emergency intubation of trauma patients. Am J Emerg Med 1997; 15:98.
  161. Kociszewski C, Thomas SH, Harrison T, Wedel SK. Etomidate versus succinylcholine for intubation in an air medical setting. Am J Emerg Med 2000; 18:757.
  162. Miner JR, Gray RO, Bahr J, et al. Randomized clinical trial of propofol versus ketamine for procedural sedation in the emergency department. Acad Emerg Med 2010; 17:604.
  163. Mortero RF, Clark LD, Tolan MM, et al. The effects of small-dose ketamine on propofol sedation: respiration, postoperative mood, perception, cognition, and pain. Anesth Analg 2001; 92:1465.
  164. Newman DH, Azer MM, Pitetti RD, Singh S. When is a patient safe for discharge after procedural sedation? The timing of adverse effect events in 1367 pediatric procedural sedations. Ann Emerg Med 2003; 42:627.
  165. Malviya S, Voepel-Lewis T, Tait AR. Adverse events and risk factors associated with the sedation of children by nonanesthesiologists. Anesth Analg 1997; 85:1207.
  166. Malviya S, Voepel-Lewis T, Prochaska G, Tait AR. Prolonged recovery and delayed side effects of sedation for diagnostic imaging studies in children. Pediatrics 2000; 105:E42.
Topic 264 Version 48.0

References

1 : Procedural sedation terminology: moving beyond "conscious sedation".

2 : Procedural sedation: providing the missing definition.

3 : Pharmacokinetic and pharmacodynamic characteristics of medications used for moderate sedation.

4 : Practice guidelines for sedation and analgesia by non-anesthesiologists.

5 : Practice guidelines for sedation and analgesia by non-anesthesiologists.

6 : Sedation and analgesia for procedures in children.

7 : Preoperative fasting for adults to prevent perioperative complications.

8 : Aspiration pneumonitis requiring intubation after procedural sedation and analgesia: a case report.

9 : The incidence and outcome of perioperative pulmonary aspiration in a university hospital: a 4-year retrospective analysis.

10 : Adverse events of emergency department procedural sedation

11 : Procedural sedation and analgesia in the emergency department: what are the risks?

12 : Side effects of opioids during short-term administration: effect of age, gender, and race.

13 : Clinical practice advisory: Emergency department procedural sedation with propofol.

14 : Pulmonary aspiration during procedural sedation: a comprehensive systematic review.

15 : Clinical policy: procedural sedation and analgesia in the emergency department.

16 : An international multidisciplinary consensus statement on fasting before procedural sedation in adults and children.

17 : Unscheduled Procedural Sedation: A Multidisciplinary Consensus Practice Guideline.

18 : Propofol for procedural sedation in children in the emergency department.

19 : Fasting and emergency department procedural sedation and analgesia: a consensus-based clinical practice advisory.

20 : Clinical practice guideline for emergency department ketamine dissociative sedation: 2011 update.

21 : Guidelines for Monitoring and Management of Pediatric Patients Before, During, and After Sedation for Diagnostic and Therapeutic Procedures: Update 2016.

22 : Effect of oral liquids and ranitidine on gastric fluid volume and pH in children undergoing outpatient surgery.

23 : Preoperative fasting for preventing perioperative complications in children.

24 : Pulmonary aspiration of gastric contents in anaesthesia.

25 : Pulmonary aspiration risk during emergency department procedural sedation--an examination of the role of fasting and sedation depth.

26 : Pre-operative fasting guidelines: an update.

27 : Sedation in children: adequacy of two-hour fasting.

28 : Profiling adverse respiratory events and vomiting when using propofol for emergency department procedural sedation.

29 : Sedation for pediatric echocardiography: evaluation of preprocedure fasting guidelines.

30 : Large volumes of apple juice preoperatively do not affect gastric pH and volume in children.

31 : Aspiration during anaesthesia: a computer-aided study of 185,358 anaesthetics.

32 : "Silent" regurgitation and aspiration during general anesthesia.

33 : Practice Guidelines for Moderate Procedural Sedation and Analgesia 2018: A Report by the American Society of Anesthesiologists Task Force on Moderate Procedural Sedation and Analgesia, the American Association of Oral and Maxillofacial Surgeons, American College of Radiology, American Dental Association, American Society of Dentist Anesthesiologists, and Society of Interventional Radiology.

34 : Clinical policy: procedural sedation and analgesia in the emergency department.

35 : Nurse-administered propofol sedation: a review of current evidence.

36 : Adverse sedation events in pediatrics: a critical incident analysis of contributing factors.

37 : The safety of single-physician procedural sedation in the emergency department.

38 : The utility of high-flow oxygen during emergency department procedural sedation and analgesia with propofol: a randomized, controlled trial.

39 : The utility of supplemental oxygen during emergency department procedural sedation and analgesia with midazolam and fentanyl: a randomized, controlled trial.

40 : Bispectral electroencephalographic analysis of patients undergoing procedural sedation in the emergency department.

41 : End-tidal carbon dioxide monitoring during procedural sedation.

42 : Capnography during sedation/analgesia in the pediatric emergency department.

43 : Supplemental oxygen during moderate sedation and the occurrence of clinically significant desaturation during endoscopic procedures.

44 : A comparative evaluation of capnometry versus pulse oximetry during procedural sedation and analgesia on room air.

45 : Capnography versus standard monitoring for emergency department procedural sedation and analgesia.

46 : Can emergency physicians recognize respiratory depression in patients who do not become hypoxic during ED procedural sedation and analgesia

47 : Does end tidal CO2 monitoring during emergency department procedural sedation and analgesia with propofol decrease the incidence of hypoxic events? A randomized, controlled trial.

48 : Conscious sedation in the emergency department: the value of capnography and pulse oximetry.

49 : A study of the Bispectral Index Monitor during procedural sedation and analgesia in the emergency department.

50 : Practice advisory for intraoperative awareness and brain function monitoring: a report by the american society of anesthesiologists task force on intraoperative awareness.

51 : Intrapatient reproducibility of the BISxp monitor.

52 : Sedation Challenges: Obesity and Sleep Apnea.

53 : Predictors of difficult airway management in morbid obesity patients

54 : Comparing ease of intubation in obese and lean patients using intubation difficulty scale.

55 : Obesity as a risk factor for sedation-related complications during propofol-mediated sedation for advanced endoscopic procedures.

56 : Pharmacotherapy of procedural sedation in morbidly obese

57 : Influence of obesity on propofol pharmacokinetics: derivation of a pharmacokinetic model.

58 : Drug dosing in the critically ill obese patient-a focus on sedation, analgesia, and delirium.

59 : Anesthetic Pharmacology and the Morbidly Obese Patient.

60 : Anesthetic Pharmacology and the Morbidly Obese Patient.

61 : Pharmacokinetics in obese patients

62 : Incidence of Adverse Events in Adults Undergoing Procedural Sedation in the Emergency Department: A Systematic Review and Meta-analysis.

63 : Clinical Practice Guideline for Emergency Department Procedural Sedation With Propofol: 2018 Update.

64 : Adverse events of procedural sedation and analgesia in a pediatric emergency department.

65 : Newer pharmacologic agents for procedural sedation of children in the emergency department-etomidate and propofol.

66 : Use of propofol sedation in a pediatric emergency department: a prospective study.

67 : Clinical policy for procedural sedation and analgesia in the emergency department. American College of Emergency Physicians.

68 : Diazepam-fentanyl interaction--hemodynamic and hormonal effects in coronary artery surgery.

69 : Pharmacology of emergency department pain management and conscious sedation.

70 : Safe and efficacious use of procedural sedation and analgesia by nonanesthesiologists in a pediatric emergency department.

71 : Etomidate for procedural sedation in emergency medicine.

72 : Recovery following tonsillectomy a comparison between tramadol and morphine for analgesia.

73 : Adverse events associated with procedural sedation and analgesia in a pediatric emergency department: a comparison of common parenteral drugs.

74 : Effect of ondansetron on the incidence of vomiting associated with ketamine sedation in children: a double-blind, randomized, placebo-controlled trial.

75 : The use of propofol for sedation in the emergency department.

76 : The use of propofol for sedation in the emergency department.

77 : Prevention of pain on injection of propofol: systematic review and meta-analysis.

78 : Small-dose ketamine reduces the pain of propofol injection.

79 : Randomized clinical trial of propofol with and without alfentanil for deep procedural sedation in the emergency department.

80 : Optimizing the Treatment of Acute Pain in the Emergency Department.

81 : Ketamine: Current applications in anesthesia, pain, and critical care.

82 : Propofol for adult procedural sedation in a UK emergency department: safety profile in 1008 cases.

83 : Propofol for procedural sedation in the emergency department: a qualitative systematic review.

84 : Nurse-administered propofol sedation without anesthesia specialists in 9152 endoscopic cases in an ambulatory surgery center.

85 : Efficacy and safety of nurse-administered propofol sedation during emergency upper endoscopy for gastrointestinal bleeding: a prospective study.

86 : The use of propofol for procedural sedation in emergency departments.

87 : Barriers to propofol use in emergency medicine.

88 : "Poachers and dabblers?": ASA president's incautious comment riles emergency physicians.

89 : The great debate on nurse-administered propofol sedation (NAPS)--where should we stand?

90 : Randomized clinical trial of etomidate versus propofol for procedural sedation in the emergency department.

91 : Procedural sedation in the acute care setting.

92 : Etomidate for procedural sedation in the emergency department.

93 : Procedural sedation in the community emergency department: initial results of the ProSCED registry.

94 : Etomidate for procedural sedation in the emergency department.

95 : Reducing myoclonus after etomidate.

96 : Etomidate and midazolam for reduction of anterior shoulder dislocation: a randomized, controlled trial.

97 : Intravenous etomidate for procedural sedation in emergency department patients.

98 : Low-dose intravenous midazolam reduces etomidate-induced myoclonus: a prospective, randomized study in patients undergoing elective cardioversion.

99 : Magnesium sulfate pretreatment reduces myoclonus after etomidate.

100 : Adrenocortical function in critically ill patients 24 h after a single dose of etomidate.

101 : Adrenocortical dysfunction following etomidate induction in emergency department patients.

102 : Pharmacology and pharmacokinetics of sedatives and analgesics.

103 : Procedural sedation and analgesia: a review and new concepts.

104 : Procedural sedation and analgesia: a review and new concepts.

105 : Remifentanil for procedural sedation: a systematic review of the literature.

106 : Remifentanil for procedural sedation in the emergency department.

107 : Evaluation of propofol and remifentanil for intravenous sedation for reducing shoulder dislocations in the emergency department.

108 : Procedural sedation and analgesia in children.

109 : Alfentanil for procedural sedation in the emergency department.

110 : Procedural sedation and analgesia in a Canadian adult tertiary care emergency department: a case series.

111 : A randomized controlled trial on procedural sedation among adult patients in emergency departments: comparing fentanyl with midazolam versus fentanyl with propofol [abstract]

112 : A randomized controlled trial on procedural sedation among adult patients in emergency departments: comparing fentanyl with midazolam versus fentanyl with propofol [abstract]

113 : Adverse events associated with ketamine for procedural sedation in adults.

114 : Intravenous ketamine for adult procedural sedation in the emergency department: a prospective cohort study.

115 : Subanesthetic doses of ketamine stimulate psychosis in schizophrenia.

116 : Ketamine-induced exacerbation of psychotic symptoms and cognitive impairment in neuroleptic-free schizophrenics.

117 : Evolution of a protocol for ketamine-induced sedation as an alternative to general anesthesia for interventional radiologic procedures in pediatric patients.

118 : A combination of midazolam and ketamine for procedural sedation and analgesia in adult emergency department patients.

119 : Audit of the safety and effectiveness of ketamine for procedural sedation in the emergency department.

120 : Ketamine with and without midazolam for emergency department sedation in adults: a randomized controlled trial.

121 : Premedication With Midazolam or Haloperidol to Prevent Recovery Agitation in Adults Undergoing Procedural Sedation With Ketamine: A Randomized Double-Blind Clinical Trial.

122 : Predictors of adverse events with intramuscular ketamine sedation in children.

123 : Ketamine safety profile in the developing world: survey of practitioners.

124 : Ketamine safety profile in the developing world: survey of practitioners.

125 : Randomized clinical trial of propofol versus methohexital for procedural sedation during fracture and dislocation reduction in the emergency department.

126 : A prospective evaluation of "ketofol" (ketamine/propofol combination) for procedural sedation and analgesia in the emergency department.

127 : Subdissociative-dose ketamine versus fentanyl for analgesia during propofol procedural sedation: a randomized clinical trial.

128 : Ketamine-Propofol Versus Propofol Alone for Procedural Sedation in the Emergency Department: A Systematic Review and Meta-analysis.

129 : Ketamine-propofol combination (ketofol) vs propofol for procedural sedation and analgesia: systematic review and meta-analysis.

130 : Research advances in procedural sedation and analgesia.

131 : Low-dose ketamine in addition to propofol for procedural sedation and analgesia in the emergency department.

132 : Low-dose ketamine in addition to propofol for procedural sedation and analgesia in the emergency department.

133 : A randomized controlled trial of ketamine/propofol versus propofol alone for emergency department procedural sedation.

134 : Propofol versus propofol/ketamine for brief painful procedures in the emergency department: clinical and bispectral index scale comparison.

135 : Randomized, double-blinded, clinical trial of propofol, 1:1 propofol/ketamine, and 4:1 propofol/ketamine for deep procedural sedation in the emergency department.

136 : Propofol or Ketofol for Procedural Sedation and Analgesia in Emergency Medicine-The POKER Study: A Randomized Double-Blind Clinical Trial.

137 : Ketamine-propofol combination (ketofol) versus propofol alone for emergency department procedural sedation and analgesia: a randomized double-blind trial.

138 : Current role of dexmedetomidine in clinical anesthesia and intensive care.

139 : Respiratory effects of dexmedetomidine in the surgical patient requiring intensive care.

140 : Dexmedetomidine pharmacodynamics: part I: crossover comparison of the respiratory effects of dexmedetomidine and remifentanil in healthy volunteers.

141 : Sedative, amnestic, and analgesic properties of small-dose dexmedetomidine infusions.

142 : The effects of increasing plasma concentrations of dexmedetomidine in humans.

143 : Upper Airway Collapsibility during Dexmedetomidine and Propofol Sedation in Healthy Volunteers: A Nonblinded Randomized Crossover Study.

144 : Effect of increasing depth of dexmedetomidine and propofol anesthesia on upper airway morphology in children and adolescents with obstructive sleep apnea.

145 : A comparison of dexmedetomidine with propofol for magnetic resonance imaging sleep studies in children.

146 : Sedation with Dexmedetomidine or Propofol Impairs Hypoxic Control of Breathing in Healthy Male Volunteers: A Nonblinded, Randomized Crossover Study.

147 : Dexmedetomodine: its use in intensive care medicine and anesthesia

148 : Dexmedetomidine use in general anaesthesia.

149 : The comparison of dexmedetomidine and midazolam used for sedation of patients during upper endoscopy: A prospective, randomized study.

150 : Dexmedetomidine sedation for laryngeal framework surgery.

151 : Initial experience with dexmedetomidine for diagnostic and interventional cardiac catheterization in children.

152 : Sole use of dexmedetomidine has limited utility for conscious sedation during outpatient colonoscopy.

153 : Dexmedetomidine-ketamine and propofol-ketamine combinations for anesthesia in spontaneously breathing pediatric patients undergoing cardiac catheterization.

154 : Dexmedetomidine and ketamine: an effective alternative for procedural sedation?

155 : Limited analgesic efficacy of nitrous oxide for painful procedures in children.

156 : The effects of speed of injection on induction with propofol. A comparison with etomidate.

157 : Propofol for emergency department procedural sedation and analgesia: a tale of three centers.

158 : Propofol sedation by emergency physicians for elective pediatric outpatient procedures.

159 : Sedation for cardioversion in the emergency department: analysis of effectiveness in four protocols.

160 : Etomidate use during emergency intubation of trauma patients.

161 : Etomidate versus succinylcholine for intubation in an air medical setting.

162 : Randomized clinical trial of propofol versus ketamine for procedural sedation in the emergency department.

163 : The effects of small-dose ketamine on propofol sedation: respiration, postoperative mood, perception, cognition, and pain.

164 : When is a patient safe for discharge after procedural sedation? The timing of adverse effect events in 1367 pediatric procedural sedations.

165 : Adverse events and risk factors associated with the sedation of children by nonanesthesiologists.

166 : Prolonged recovery and delayed side effects of sedation for diagnostic imaging studies in children.