Your activity: 6 p.v.

Ketamine: Pediatric drug information

Ketamine: Pediatric drug information
(For additional information see "Ketamine: Drug information" and see "Ketamine: Patient drug information")

For abbreviations, symbols, and age group definitions used in Lexicomp (show table)
Brand Names: US
  • Ketalar
Brand Names: Canada
  • Ketalar
Therapeutic Category
  • General Anesthetic
Dosing: Neonatal

Note: Individualize dose and titrate to effect. Some neonatal experts do not recommend the use of ketamine in neonates; an increase in neuronal apoptosis has been observed in neonatal animal studies (Ref). The American College of Emergency Physicians considers the use of ketamine in infants <3 months of age to be an absolute contraindication, due to the higher risk of airway complications (Ref).

Anesthesia, adjunct

Anesthesia, adjunct: Limited data available: IV: 0.5 to 2 mg/kg/dose has been reported. A study of 23 patients [mean age: 3.2 years (9 days to 7 years)] undergoing MRI received a fixed dose of 0.5 mg/kg of ketamine prior to propofol bolus and infusion. A single case report describes ketamine (1 to 2 mg/kg) adjunct use with sevoflurane in a 1-day old neonate (GA: 33 weeks) undergoing pacemaker placement (Ref).

Procedural sedation/analgesia

Procedural sedation/analgesia: Limited data available: IV: 0.5 to 2 mg/kg/dose (Ref); reported use includes intubation, during cardiac catheterization, and ROP corrective procedures (Ref).

Dosing: Pediatric

Note: Titrate dose to effect. May be used in combination with anticholinergic agents to decrease hypersalivation. Note: The American College of Emergency Physicians considers the use of ketamine in infants <3 months of age to be an absolute contraindication, due to the higher risk of airway complications (Ref).

Analgesia, acute pain

Analgesia, acute pain (low dose; sub-dissociative): Very limited data available: Optimal dose not established: Note: Use of mucosal atomizer device is recommended:

Children ≥3 years and Adolescents: Intranasal: Usual: 1 mg/kg/dose, may repeat once; range: 0.5 to 1.5 mg/kg/dose; maximum dose: 100 mg/dose (Ref). Dosing based on 2 double-blind, randomized, controlled studies (n=80 in ketamine treatment group, age range: 3 to 17 years) comparing intranasal ketamine to intranasal fentanyl in the treatment of acute pain due to isolated limb injury or suspected single extremity fracture in the emergency department; pain relief was similar between both treatment groups; no serious adverse effects were reported (Ref).

Anesthesia

Anesthesia:

Pre-anesthetic sedation: Limited data available:

Intranasal:

Infants ≥6 months: 3 mg/kg/dose (half dose per nostril) administered at least 15 minutes prior to mask induction (Ref).

Children <2 years: 3 to 5 mg/kg/dose (half dose per nostril) administered at least 15 minutes prior to mask induction (Ref).

Children 2 to 7 years: 3 to 6 mg/kg/dose (half dose per nostril) administered 15 to 40 minutes prior to induction (Ref).

Oral: Children ≤8 years: 6 to 8 mg/kg/dose 20 to 30 minutes prior to surgery. Dosing based on 2 prospective, randomized, double-blind, placebo-controlled, dose finding trials. The larger trial compared 4 mg/kg, 6 mg/kg, and 8 mg/kg (n=20 in each group; age range: 2 to 8 years) and found that patients who received 8 mg/kg were significantly more calm, but also had slightly longer recovery times. Sedation was effective within 10 minutes of administration in 80% and 45% of the patients in the 8 mg/kg and 6 mg/kg, groups respectively, but was not effective in the 4 mg/kg group (Ref). The other study compared 3 mg/kg and 6 mg/kg (n=15 in each group, age range: 1 to 7 years) and found that patients who received 6 mg/kg had satisfactory sedation without prolonged recovery times, but the 3 mg/kg dose did not provide uniform sedation nor offer a significant improvement in premedicated emotional state compared to placebo (Ref).

Rectal: Administer 15 to 45 minutes prior to surgery as a single agent; when used in combination with other sedatives, lower doses should be considered. Efficacy was reported in trials comparing rectal ketamine to rectal doses of other agents (fentanyl/droperidol, midazolam) (Ref). Reported effective range:

Infants 2 to 6 months: 8 mg/kg/dose.

Infants ≥7 months and Children ≤9 years: 8 to 10 mg/kg/dose.

Note: Although lower doses of 4 to 7 mg/kg/dose have been reported, they were less effective. In some patients, the 10 mg/kg/dose was associated with prolonged postoperative sedation. When used in combination with midazolam, a lower rectal dose of 3 mg/kg/dose has been effective (Ref).

Induction of anesthesia:

Infants ≥3 months, Children, and Adolescents <16 years: Limited data available:

IM: 5 to 10 mg/kg has been reported and suggested by experts (Ref).

IV: 1 to 3 mg/kg has been reported and suggested by experts (Ref).

Adolescents ≥16 years:

IM: 6.5 to 13 mg/kg.

IV: 1 to 4.5 mg/kg.

Maintenance of anesthesia: Adolescents ≥16 years: May administer supplemental doses of one-half to the full induction dose as needed.

Endotracheal intubation

Endotracheal intubation: Limited data available: Infants, Children, and Adolescents: IV: 1 to 2 mg/kg as part of rapid sequence sedation (Ref).

Sedation/analgesia, procedural

Sedation/analgesia, procedural: Limited data available:

Ketamine without propofol: Infants ≥3 months, Children, and Adolescents:

IM: 4 to 5 mg/kg as a single dose; may give a repeat dose (range: 2 to 5 mg/kg) if sedation inadequate after 5 to 10 minutes or if additional doses are required (Ref). Some have recommended smaller doses (2 to 2.5 mg/kg) for minor procedures (eg, wound suture with local anesthetic) (Ref).

IV: 1 to 2 mg/kg over 30 to 60 seconds. If initial sedation inadequate or repeated doses are necessary to accomplish a longer procedure, may administer additional doses of 0.5 to 1 mg/kg every 5 to 15 minutes as needed (Ref).

Intranasal: Note: Use of mucosal atomizer device is recommended:

Infants ≥3 months and Children: 3 to 6 mg/kg (half dose per nostril), doses up to 9 mg/kg have been described (Ref).

Oral: Children and Adolescents: 5 mg/kg with oral midazolam given 30 to 45 minutes before the procedure. Dosing based on 2 prospective, randomized, blinded studies involving patients aged 1 to 10 years with either laceration repair or burn wound care (Ref). A lower dose (3 mg/kg) in addition to midazolam was effective in a study comparing different routes (IV, oral, rectal) of ketamine plus midazolam for invasive procedures in oncology patients. The oral group included 24 patients (mean age: 3.9 ± 1.3 years); incidence of optimal sedation was similar between groups (75% for the oral group) (Ref). Note: A higher dose (10 mg/kg) has been used successfully as monotherapy prior to procedures in pediatric oncology patients (n=35, age: 14 months to 17 years; mean age: 6.5 years) (Ref).

Rectal: Children 1 to 8 years: 1.5 to 3 mg/kg with midazolam as a single dose 20 minutes prior to painful procedure. Dosing based on 2 studies. The first was completed in children with burns requiring dressing changes (n=47 procedures in 30 patients, mean age: 1.9 years, range: 10 months to 7.3 years). Patients received 0.75 mg/kg of the S(+) isomer (equivalent to 1.5 mg/kg of racemic ketamine) along with rectal midazolam 20 minutes prior to dressing changes; 94% of the procedures were reported to have good or excellent analgesia (Ref). The second study compared different routes (IV, oral, rectal) of ketamine plus midazolam for invasive procedures in oncology patients. The rectal group included 24 patients (mean age: 3.7 ± 1.1 years) who received 3 mg/kg of ketamine in addition to midazolam. Incidence of optimal sedation was similar between groups (79% for the rectal group) (Ref). In both of these studies, rectal ketamine was well tolerated.

Ketamine with propofol ("ketofol"): Infants ≥3 months, Children, and Adolescents: IV: 0.5 to 0.75 mg/kg of each agent. This combination has been used to decrease the dose of each agent required. It has been proposed that these lower doses help decrease adverse effects, ketamine may decrease the propofol-related hypotension and respiratory depression, and propofol may decrease the ketamine associated nausea and emergence reactions (Ref).

Sedation/analgesia, critically ill patients

Sedation/analgesia, critically ill patients: Very limited data available: Infants ≥5 months, Children, and Adolescents: Initial dose: IV: 0.5 to 2 mg/kg, then continuous IV infusion: 5 to 20 mcg/kg/minute (0.3 to 1.2 mg/kg/hour); start at lower dosage listed and titrate to effect (Ref); doses as high as 60 mcg/kg/minute (3.6 mg/kg/hour) have been reported in patients with refractory bronchospasm (Ref).

Dosage adjustment for concomitant therapy: Significant drug interactions exist, requiring dose/frequency adjustment or avoidance. Consult drug interactions database for more information.

Dosing: Kidney Impairment: Pediatric

There are no dosage adjustments provided in the manufacturer's labeling.

Dosing: Hepatic Impairment: Pediatric

There are no dosage adjustments provided in the manufacturer's labeling.

Dosing: Adult

(For additional information see "Ketamine: Drug information")

Note: Safety: To decrease risk of respiratory depression and apnea, administer IV bolus doses over >30 to 60 seconds (Ref).

Agitation, acute/severe or refractory

Agitation, acute/severe or refractory (adjunct) (off-label use): Note: May be used when benzodiazepines and/or antipsychotics have failed (Ref).

IV: Initial: 1 to 2 mg/kg once over 30 to 60 seconds; if initial sedation is inadequate, may repeat dose once 5 to 10 minutes after the initial dose using 0.5 to 1 mg/kg; however, this is not common if using the upper end of the initial dose range (Ref). Some experts use an initial dose of 0.5 to 1 mg/kg in the emergency department with concomitant sedatives (Ref).

IM: Initial: 4 to 6 mg/kg once; if initial sedation is inadequate, may repeat dose once 10 to 25 minutes after the initial dose using 2 to 3 mg/kg; however, this is not common if using the upper end of the initial dose range (Ref). Some experts use an initial dose of 2 mg/kg in the emergency department with concomitant sedatives (Ref).

Analgesia, subanesthetic dosing

Analgesia, subanesthetic dosing (off-label use):

Acute pain:

Note: Prior to use, consult with a pain specialist or service experienced with ketamine use in this setting. Optimal doses and regimens have not been identified; refer to institutional protocols. Recommendations provided below are examples of regimens. May be useful for moderately to severely painful procedures and conditions that do not respond optimally to standard analgesics (eg, postoperative, burn, trauma, sickle cell disease vaso-occlusive pain).

IV: Initial: 0.25 to 0.5 mg/kg bolus (maximum bolus: 35 mg), followed by 0.05 to 0.25 mg/kg/hour continuous infusion in patients who need a longer duration of analgesia; titrate to pain goal and tolerability; usual dosing range: 0.05 to 1 mg/kg/hour; may need to use doses at the higher range in patients who are opioid-tolerant or with opioid-induced hyperalgesia; duration of infusion: 48 to 72 hours (Ref).

Intranasal (off-label route): 0.2 to 1 mg/kg by administering half dose in each nostril (using 100 mg/mL solution); if necessary, may repeat after 10 to 15 minutes with 0.25 to 0.5 mg/kg; titrate to pain goal and tolerability. Doses up to 40 mg may be reliably administered intranasally; for doses >40 mg, part of the dose will be delivered to the oropharynx and ingested orally due to volume limitations, which may decrease effectiveness (Ref).

Chronic pain, intractable:

Note: Prior to use, consult with a pain specialist or service experienced with ketamine use in this setting. Optimal regimens, doses, and duration have not been identified; refer to institutional protocols. Long-term safety, efficacy, and timing of repeated ketamine treatments are not well established. Recommendations provided below are examples of regimens. May be used in refractory chronic pain (eg, complex regional pain syndrome, end-stage illness, neuropathies). May need to use doses at the higher range in opioid-tolerant patients (Ref). Reduce baseline opioids by 25% to 50% when used concomitantly with ketamine (Ref).

IV intermittent infusion: Initial: 0.25 to 0.6 mg/kg (usual maximum dose: 60 mg) as a 4- to 6-hour infusion; titrate to pain goal and tolerability; repeat daily for up to 2 to 10 days as needed (Ref).

IV continuous infusion: Initial: 0.05 to 0.15 mg/kg/hour for 1 day outpatient or for 2 to 5 days inpatient; titrate to pain goal and tolerability; usual dosing range: 0.02 to 1 mg/kg/hour (maximum dose: 30 mg/hour; not well established) (Ref).

SubQ: Initial: 0.1 to 0.6 mg/kg (usual 2.5 to 25 mg) as needed; titrate to pain goal and tolerability. In patients who need a longer duration of analgesia, follow with continuous SubQ infusion at 0.1 to 1.2 mg/kg/hour (maximum daily dose: 500 mg) (Ref).

Oral: Note: Used in refractory chronic pain (eg, advanced illness or palliative care) in a hospitalized patient when other regimens have failed.

Initial: 0.5 mg/kg/day administered in 3 to 4 divided doses as needed; then increase dose in increments of ~5 mg/dose based on pain goal and tolerability; maximum daily escalation dose: 15 to 20 mg; maximum dose: 800 mg/day (Ref). Note: May administer dose as the undiluted injectable or mixed with an appropriate flavoring agent (eg, simple syrup).

Depressive episode associated with major depressive disorder

Depressive episode (severe, treatment resistant) associated with major depressive disorder (unipolar) (off-label use): Note: Avoid or use with caution in patients with substance use disorder.

IV: Initial: 0.5 mg/kg administered over 40 minutes; may repeat at a frequency of 1 to 3 times weekly; may increase dose to 0.75 to 1 mg/kg based on response and tolerability. Treatment up to 6 weeks has been studied, although optimal duration of therapy is unknown (Ref). In those who respond to short-term ketamine infusions (eg, 1 to 4 weeks), some experts recommend transitioning to maintenance treatment with antidepressants and/or psychotherapy instead of prolonged ketamine treatment (Ref).

General anesthesia

General anesthesia (alternative agent):

Induction of anesthesia:

Note: Useful in hypotensive patients or patients likely to develop hypotension during induction; lower doses may be used if concomitant anesthesia/sedatives (eg, midazolam) are administered (Ref).

IV: 0.5 to 2 mg/kg or 0.5 to 1 mg/kg in patients with shock (Ref).

IM (use only if IV access is not available): 4 to 6 mg/kg (Ref).

Maintenance of anesthesia (adjunct with total intravenous anesthesia): Note: Adjunct to reduce requirements of other anesthetic agents. Typically reserved for opioid-tolerant patients in order to avoid or limit opioid requirements (Ref); concurrent use of nitrous oxide reduces ketamine requirements (Ref).

IV: 0.25 to 0.35 mg/kg, followed by continuous infusion up to 1 mg/kg/hour (Ref).

Mechanically ventilated patients in the ICU, analgesia/sedation/agitation

Mechanically ventilated patients in the ICU, analgesia/sedation/agitation (adjunct or alternative agent) (off-label use):

Note: Used as part of a multimodal strategy and adjunct for the reduction of opioid and sedative requirements and/or for the management of opioid-induced hyperalgesia in patients with acute/chronic pain or postsurgical pain. Pain should be monitored using validated scales (eg, behavioral pain scale, critical-care pain observation tool) in patients who are unable to self-report. If used for sedation, titrate to a light level of sedation (eg, Richmond Agitation Sedation Scale 0 to −2), unless deeper sedation is clinically indicated (Ref). Refer to institutional policies and procedures.

IV: Initial: 0.1 to 0.5 mg/kg bolus, followed by a 0.2 to 0.5 mg/kg/hour continuous infusion; titrate dose to pain and/or sedation goal (dosing range: 0.04 to 2.5 mg/kg/hour) (Ref). Note: Higher initial bolus doses (eg, 1 to 2 mg/kg) can be used to manage severely agitated patients; see "Agitation, acute/severe or refractory (off-label use)" (Ref).

Procedural sedation

Procedural sedation (off-label use): Note: To decrease risk of emergence reactions (although rare), may consider premedication with a benzodiazepine (eg, midazolam) (Ref).

IV: Initial: 1 to 2 mg/kg over 1 to 2 minutes; if initial sedation is inadequate or for longer procedures, repeat dose (0.5 to 1 mg/kg) every 5 to 10 minutes; use lower doses (0.25 to 0.5 mg/kg) depending on concomitant sedation and clinical status (Ref). Some experts use 0.5 to 0.75 mg/kg (as a 1:1 mixture) when combined with propofol (Ref).

IM: Note: Onset of sedation will be delayed ~5 minutes with this route.

Initial: 4 to 5 mg/kg as a single dose; if sedation is inadequate after 5 to 10 minutes, repeat dose (2 to 5 mg/kg) (Ref).

Rapid sequence intubation outside the operating room

Rapid sequence intubation outside the operating room (induction) (off-label use): Note: Consider in patients with bronchospasm and/or hemodynamic compromise (Ref).

IV: Initial: 1 to 2 mg/kg once over 1 minute; in patients with shock, use 1 mg/kg (Ref).

Intraosseous (use only if IV access is not available): Initial: 100 mg once (Ref).

Status epilepticus, refractory

Status epilepticus, refractory (off-label use): Note: Used as an alternative or adjunct to midazolam, propofol, or barbiturates after conventional intermittent antiseizure therapies have failed. Mechanical ventilation and hemodynamic support generally required; continuous EEG is recommended; titrate doses to cessation of electrographic seizures or burst suppression (Ref). Optimal regimen and dose are uncertain; refer to institutional protocol.

IV:

Loading dose: Initial: 1.5 mg/kg or 0.5 to 3 mg/kg; repeat loading dose of 0.5 mg/kg every 3 to 5 minutes as needed for electrographic/burst suppression, followed by continuous infusion (Ref).

Continuous infusion: After initial loading dose and electrographic suppression, begin at a rate of 0.1 to 4 mg/kg/hour; titrate as needed for electrographic/burst suppression; maximum dose: 15 mg/kg/hour (Ref).

Note: Generally, a period of at least 24 hours of electrographic suppression is suggested prior to down-titrating the continuous infusion; withdraw gradually by decreasing the dose by 20% every 3 hours while continuing conventional antiseizure therapies (Ref).

Dosage adjustment for concomitant therapy: Significant drug interactions exist, requiring dose/frequency adjustment or avoidance. Consult drug interactions database for more information.

Dosing: Kidney Impairment: Adult

There are no dosage adjustments provided in the manufacturer's labeling.

Dosing: Hepatic Impairment: Adult

There are no dosage adjustments provided in the manufacturer's labeling.

Dosage Forms: US

Excipient information presented when available (limited, particularly for generics); consult specific product labeling.

Solution, Injection:

Ketalar: 10 mg/mL (20 mL); 50 mg/mL (10 mL); 100 mg/mL (5 mL)

Generic: 10 mg/mL (20 mL); 50 mg/mL (10 mL); 100 mg/mL (5 mL, 10 mL)

Solution Prefilled Syringe, Intravenous, as hydrochloride:

Generic: 50 mg/5 mL (5 mL)

Generic Equivalent Available: US

Yes

Dosage Forms: Canada

Excipient information presented when available (limited, particularly for generics); consult specific product labeling.

Solution, Injection:

Ketalar: 10 mg/mL (10 mL, 20 mL, 50 mL); 50 mg/mL (10 mL) [contains benzethonium chloride]

Generic: 10 mg/mL (2 mL, 20 mL); 50 mg/mL (2 mL, 10 mL)

Controlled Substance

C-III

Administration: Pediatric

Intranasal (using parenteral dosage form): Use the 50 or 100 mg/mL solution; may administer undiluted or further diluted in NS to a concentration of 20 mg/mL (Ref). Administer dose in 1 nostril (for volumes ≤0.5 mL) or divide dose and administer half dose in each nostril using a mucosal atomizer device (preferred) or needleless syringe (Ref).

Oral (using parenteral dosage form): Use the 100 mg/mL IV solution and mix the appropriate dose in 0.2 to 0.4 mL/kg of cola, sour cherry juice, or other beverage (Ref).

Parenteral:

IV push: May administer the 10 mg/mL and 50 mg/mL undiluted. Administer slowly over 60 seconds, do not exceed 0.5 mg/kg/minute; more rapid administration may result in respiratory depression and enhanced pressor response. Some experts suggest administration over 2 to 3 minutes (Ref).

IV infusion: May be administered as a continuous IV infusion.

Rectal (using parenteral dosage form): May use the 50 mg/mL solution undiluted or use the 100 mg/mL solution and further dilute prior to administration (Ref).

Administration: Adult

Intranasal (off label): Administer half dose in each nostril using a needleless syringe or mucosal atomizer device. Note: Intranasal use for adults for procedural sedation is not recommended since volume limits an adequate dose (Ref).

Oral (off label): May administer undiluted or mix the appropriate dose (using the injectable solution) in a flavoring agent (eg, simple syrup), cola, or other beverage; administer immediately after preparation (Ref).

IM: Inject deep IM into large muscle mass.

IV: According to the manufacturer, administer bolus/induction doses over 1 minute or at a rate of 0.5 mg/kg/minute; more rapid administration may result in respiratory depression and enhanced pressor response. Some experts suggest administration over 2 to 3 minutes (Ref). When used for treatment refractory unipolar depression, administer over 40 minutes (Ref). May also be administered as a continuous infusion.

Rectal (off label): May use the 50 mg/mL solution undiluted or use the 100 mg/mL solution and further dilute prior to administration (Ref).

SubQ (off label): May administer as a subcutaneous continuous infusion (Ref).

Storage/Stability

Vials: Store intact vials at 20°C to 25°C (68°F to 77°F); excursions permitted to 15°C to 30°C (59°F to 86°F). Protect from light.

Oral solution (off label): Store prepared oral solution at 2°C to 8°C (36°F to 46°F) for up to 1 week from preparation (Quibell 2015).

Use

Induction and maintenance of general anesthesia (FDA approved in ages ≥16 years and adults); has also been used for the treatment of acute pain/analgesia, procedural sedation/analgesia (including endotracheal intubation), and sedation of critically ill patients.

Medication Safety Issues
Sound-alike/look-alike issues:

Ketalar may be confused with Kenalog, ketorolac

High alert medication:

The Institute for Safe Medication Practices (ISMP) includes this medication (IV, intranasal/inhaled administration, parenteral used orally for sedation in children) among its list of drugs which have a heightened risk of causing significant patient harm when used in error.

Adverse Reactions (Significant): Considerations
Airway complications

When used for procedural sedation for major procedures involving the posterior pharynx (eg, endoscopy) or in patients with an active pulmonary infection or disease (including upper respiratory disease or asthma), the use of ketamine increases the risk of laryngospasm (rare and typically transient) (Ref). The manufacturer recommends against the use of ketamine alone in surgery or diagnostic procedures of the pharynx, larynx, or bronchial tree; mechanical stimulation of the pharynx should be avoided, whenever possible, if ketamine is used alone.

Mechanism: Dose-related; ketamine does not suppress pharyngeal and laryngeal reflexes. Laryngospasm may occur secondary to stimulation of the vocal cords by instrumentation and/or ketamine-induced increased salivary secretions (Ref).

Risk factors:

• Higher IV doses (Ref)

• IM administration (and higher doses) (Ref)

• Pediatric patients <2 years or ≥13 years of age (Ref)

• Active pulmonary infection or disease, including upper respiratory disease or asthma (Ref)

• History of airway instability, tracheal surgery, or tracheal stenosis (Ref)

Cardiovascular effects

Ketamine causes increased blood pressure, increased heart rate, and increased cardiac output, thereby increasing myocardial oxygen demand; cardiac arrhythmia, cardiac decompensation, decreased blood pressure, and decreased heart rate have also been reported (Ref). In a scientific statement from the American Heart Association, ketamine has been determined to be an agent that may exacerbate underlying myocardial dysfunction (magnitude: major) (Ref). Resolution of increased blood pressure usually occurs within 15 minutes after the peak.

Mechanism: Ketamine has both central sympathetic stimulation and inhibition of catecholamine uptake and direct negative inotropic effects. Central sympathetic stimulation results in increased blood pressure, heart rate, cardiac output, and therefore, increased myocardial oxygen demand (Ref). Direct negative inotropic effects may predominate in patients with heart failure and contribute to decompensation (Ref).

Onset: Rapid; a peak in blood pressure occurs within a few minutes of administration.

Risk factors:

• Known or suspected coronary artery disease, angina, hypertension (Ref)

• Older adults with risk factors for coronary artery disease (Ref)

• Heart failure (Ref)

• Catecholamine depletion (hypotension) (Ref)

Dependence

Ketamine may cause drug dependence (withdrawal symptoms on discontinuation) and drug tolerance with prolonged use (Ref). Multiple case reports of ketamine misuse have been described (Ref). A withdrawal syndrome following discontinuation may include symptoms such as shaking, sweating, palpitations, fatigue, decreased appetite, chills, autonomic arousal, lacrimation, restlessness, cravings, dysphoria, anxiety, depressed mood, nightmares, paranoia, delusions, and hallucinations (Ref). Typical duration of withdrawal symptoms is ~3 days, with some cases persisting for 2 weeks (Ref).

Mechanism: Dose- and time-related; development of dependence may be related to the psychological effects of ketamine and tolerance. Ketamine is structurally similar to phencyclidine and the psychological effects contributing to dependence may include euphoria, perceptual changes, dissociation, and hallucinations. Tolerance most likely occurs due to auto-induction of metabolism (Ref). Dependence may also be related to N-methyl-D-aspartate (NMDA) glutamate receptor blockade, producing alcohol-like subjective effects, as well as opioid effects (Ref).

Onset: Dependence: Delayed; from isolated case reports, dependence may evolve over weeks to years (Ref). Withdrawal syndrome: Rapid; symptoms usually occur within 24 hours of discontinuation (Ref).

Risk factors:

• Long-term high-dose use (Ref)

• Rapid dose escalation (Ref)

• History of multiple substance use disorders (eg, alcohol, cannabis, opioids) (Ref)

• Ease of access (Ref)

Emergence reactions

Prolonged emergence from anesthesia, which can manifest as vivid dreams, hallucinations, and/or frank delirium commonly occur in all ages (Ref).

Mechanism: Dose-related; may occur due to depression of auditory and visual relay nuclei, leading to misperception and/or misinterpretation of auditory and visual stimuli (Ref).

Onset: Rapid; emergence reactions may occur up to 24 hours postoperatively.

Risk factors:

• Higher doses (Ref)

• Rapid IV administration (Ref)

• Age >16 years (Ref)

• Females (Ref)

• Excessive noise or stimulation during recovery (Ref)

• People who normally dream or have a history of a personality disorder (Ref)

• Ketamine monotherapy (concurrent use with a benzodiazepine may reduce risk) (Ref)

Genitourinary effects

Lower urinary tract and bladder symptoms, including dysuria, urinary frequency, urinary urgency, urinary incontinence, hematuria, and nocturia, have been reported in patients with a history of chronic ketamine use or abuse and may be related to ketamine treatment, not the underlying condition. Additional findings from diagnostic studies have included cystitis, hydronephrosis, and bladder dysfunction (reduced capacity) (Ref). Reversibility of symptoms depends on time to diagnosis and intervention (Ref).

Mechanism: Not clearly established; several proposed mechanisms, including direct effects of ketamine and the active metabolite, norketamine/hydroxynorketamine on bladder mucosa (Ref).

Onset: Delayed; case reports range from 1 month to 4.5 years (Ref).

Risk factors:

• Chronic use or abuse (Ref)

• Frequency of use of other drugs (eg, opiates, methamphetamine) (Ref)

• Female gender (Ref)

• History of urinary tract infections (Ref)

Neurodevelopmental effects in children

Animal studies have shown that prolonged or repeated exposure to medications for anesthesia or sedation cause adverse effects on brain maturation resulting in changes in behavior and learning. Some human studies have also suggested similar effects, including epidemiological studies in humans that have observed various cognitive and behavioral problems, including neurodevelopmental delay (and related diagnoses), learning disabilities, and attention deficit hyperactivity disorder. However, data are limited, inconclusive, and further studies are needed to fully characterize findings. Based on the potential risk, the FDA warned in 2016 that in neonates, infants, children <3 years, and patients in the third trimester of pregnancy (ie, times of rapid brain growth and synaptogenesis) undergoing repeated or lengthy exposure to sedatives or anesthetics during surgery/procedures/critical illness, there may be detrimental effects on the child's or fetus’ brain development which may lead to various cognitive and behavioral problems. Relatively short exposure (<3 hours) to sedatives or anesthetics during surgery or procedures is unlikely to adversely affect brain development (Ref).

Mechanism: Unknown; in juvenile animal studies, drugs that potentiate gamma-aminobutyric acid activity and/or block N-methyl-D aspartate receptors for >3 hours demonstrated widespread neuronal and oligodendrocyte cell loss along with alteration in synaptic morphology and neurogenesis (Ref).

Risk factors:

• Neonates, infants, children <3 years, and pregnant patients in their third trimester undergoing procedures lasting >3 hours or multiple procedures (Ref)

Respiratory depression

Ketamine may rarely cause respiratory depression or apnea, which are usually transient. These adverse reactions are most associated with rapid IV administration but have also been reported with IM administration (Ref). According to the manufacturer, overdose is also a risk factor for these adverse reactions; however, respiratory depression and apnea have been reported in the setting of slow sub-anesthetic IV infusion (Ref).

Onset: Rapid; typically, 1 to 2 minutes after IV administration or 4 to 5 minutes after IM administration (Ref). In the case of respiratory depression and apnea in the setting of slow sub-clinical IV infusion, onset occurred 25 minutes after the start of ketamine infusion (Ref).

Risk factors:

• Overdose

• Rapid IV administration (Ref)

• Concurrent use of other CNS depressants (eg, benzodiazepines, opioids)

Adverse Reactions

The following adverse drug reactions and incidences are derived from product labeling unless otherwise specified.

>10%: Nervous system: Prolonged emergence from anesthesia (12%; includes agitation, confusion, delirium, dreamlike state, excitement, hallucinations, irrational behavior, vivid imagery)

Frequency not defined:

Cardiovascular: Cardiac arrhythmia, cardiac decompensation, decreased blood pressure, decreased heart rate, increased blood pressure, increased heart rate

Dermatologic: Erythema of skin, morbilliform rash

Gastrointestinal: Anorexia, nausea, vomiting

Local: Pain at injection site, rash at injection site

Nervous system: Hypertonia (tonic-clonic movements sometimes resembling seizures), psychiatric disturbance

Ophthalmic: Diplopia, increased intraocular pressure, nystagmus disorder

Respiratory: Airway obstruction

Postmarketing:

Cardiovascular: Ventricular premature contractions (Cabbabe 1985)

Endocrine & metabolic: Central diabetes insipidus (Hatab 2014)

Gastrointestinal: Biliary tract disease (dilation; Cotter 2021), cholangitis (Cotter 2021), sialorrhea (Hatab 2014)

Genitourinary: Bladder dysfunction (reduced capacity) (Wei 2013), cystitis (including cystitis noninfective, cystitis interstitial, cystitis ulcerative, cystitis erosive, cystitis hemorrhagic) (Wei 2013), dysuria (Wei 2013), hematuria (Wei 2013), nocturia (Wei 2013), urinary frequency (Wei 2013), urinary incontinence (Wei 2013), urinary urgency (Wei 2013)

Hepatic: Abnormal hepatobiliary function (with recurrent or continuous use; including pericholeductal fibrosis) (Cotter 2021), hepatic cirrhosis (Cotter 2021), increased direct serum bilirubin (Cotter 2021), increased gamma-glutamyl transferase (Cotter 2021), increased liver enzymes (Cotter 2021), increased serum alanine aminotransferase (Cotter 2021), increased serum alkaline phosphatase (Cotter 2021), increased serum aspartate aminotransferase (Cotter 2021), increased serum bilirubin (Cotter 2021), jaundice (Cotter 2021)

Hypersensitivity: Anaphylaxis (Chow 2021)

Nervous system: Drug dependence (Pal 2002), increased cerebrospinal fluid pressure (List 1972), withdrawal syndrome (Cosci 2020)

Neuromuscular & skeletal: Laryngospasm (Green 2011)

Renal: Hydronephrosis (Wei 2013)

Respiratory: Apnea (Driver 2017; Gómez-Revuelta 2020), respiratory depression (Melendez 2009)

Miscellaneous: Drug tolerance

Contraindications

Hypersensitivity to ketamine or any component of the formulation; conditions in which an increase in blood pressure would be hazardous

Note: When used for procedural sedation and analgesia in the emergency department, the following additional absolute contraindications according to the American College of Emergency Physicians have been asserted (ACEP [Green 2011]): Infants <3 months of age; known or suspected schizophrenia (even if currently stable or controlled with medications)

Canadian labeling: Additional contraindications (not in US labeling): History of cerebrovascular accident; severe cardiac decompensation; surgery of the pharynx, larynx, or bronchial tree unless adequate muscle relaxants are used

Warnings/Precautions

Concerns related to adverse effects:

• CNS depression: May cause CNS depression, which may impair physical or mental abilities; patients must be cautioned about performing tasks that require mental alertness (eg, operating machinery, driving). When used for outpatient surgery, the patient should be accompanied by a responsible adult. Driving, operating hazardous machinery, or engaging in hazardous activities should not be undertaken for ≥24 hours after anesthesia, according to the manufacturer.

• Increased intracranial pressure: Some consider the use of ketamine in patients with CNS masses, CNS abnormalities, or hydrocephalus a relative contraindication due to multiple reports that ketamine may increase intracranial pressure in these patients; use caution, especially at higher doses (ACEP [Green 2011]; Cohen 2018). However, assuming adequate ventilation, some evidence suggests that ketamine has minimal effects on intracranial pressure and may even improve cerebral perfusion and reduce intracranial pressure (Albanese 1997; Bowles 2012; Quibell 2015; Zeiler 2014).

• Increased ocular pressure: Use with caution in patients with increased intraocular pressure (IOP). Some recommend avoiding use in patients with an open eye injury or other ophthalmologic disorder where an increase in IOP would prove to be detrimental; however, the effects of ketamine on IOP is mixed with some evidence demonstrating no clinically significant effect on IOP (ACEP [Green 2011]; Cunningham 1986; Drayna 2012; Miller 2010; Nagdeve 2006).

• Liver injury: Recurrent use (eg, abuse/misuse, medically supervised unapproved use) may cause hepatobiliary dysfunction (usually a cholestatic pattern) and biliary duct dilatation with or without evidence of biliary obstruction.

• Porphyria: The ACEP considers the use of ketamine in patients with porphyria a relative contraindication due to enhanced sympathomimetic effect produced by ketamine (ACEP [Green 2011]).

• Thyroid disorders: The ACEP considers the use of ketamine in patients with a thyroid disorder or receiving a thyroid medication a relative contraindication due to enhanced sympathomimetic effect produced by ketamine (ACEP [Green 2011]).

Disease-related concerns:

• Cerebrospinal fluid pressure elevation: Use with caution in patients with cerebrospinal fluid (CSF) pressure elevation; an increase in CSF pressure may be associated with use.

• Ethanol use: Use with caution in the chronic alcoholic or acutely alcohol-intoxicated.

Special populations:

• Pediatric neurotoxicity: In pediatric and neonatal patients <3 years of age and patients in third trimester of pregnancy (ie, times of rapid brain growth and synaptogenesis), the repeated or lengthy exposure to sedatives or anesthetics during surgery/procedures may have detrimental effects on child or fetal brain development and may contribute to various cognitive and behavioral problems. Epidemiological studies in humans have reported various cognitive and behavioral problems, including neurodevelopmental delay (and related diagnoses), learning disabilities, and attention deficit hyperactivity disorder. Human clinical data suggest that single, relatively short exposures are not likely to have similar negative effects. No specific anesthetic/sedative has been found to be safer. For elective procedures, risk versus benefits should be evaluated and discussed with parents/caregivers/patients; critical surgeries should not be delayed (FDA 2016).

Other warnings/precautions:

• Experienced personnel: Use requires careful patient monitoring, should only be used by experienced personnel who are not actively engaged in the procedure or surgery. If used in a nonintubated and/or nonmechanically ventilated patient, qualified personnel and appropriate equipment for rapid institution of respiratory and/or cardiovascular support must be immediately available. Use to induce moderate (conscious) sedation in patients warrants monitoring equivalent to that seen with deep anesthesia. Consult local regulations and individual institutional policies and procedures.

Warnings: Additional Pediatric Considerations

Postanesthetic emergence reactions may be minimized by limiting verbal, tactile, and visual patient stimulation during recovery, or by pretreatment with a benzodiazepine (using lower recommended doses of ketamine). Severe emergent reactions may require treatment with a small hypnotic dose of a short or ultrashort acting barbiturate.

Metabolism/Transport Effects

Substrate of CYP2B6 (major), CYP2C9 (minor), CYP3A4 (major); Note: Assignment of Major/Minor substrate status based on clinically relevant drug interaction potential

Drug Interactions

Note: Interacting drugs may not be individually listed below if they are part of a group interaction (eg, individual drugs within “CYP3A4 Inducers [Strong]” are NOT listed). For a complete list of drug interactions by individual drug name and detailed management recommendations, use the Lexicomp drug interactions program by clicking on the “Launch drug interactions program” link above.

Note: Interacting drugs may not be individually listed below if they are part of a group interaction (eg, individual drugs within “CYP3A4 Inducers [Strong]” are NOT listed). For a complete list of drug interactions by individual drug name and detailed management recommendations, use the Lexicomp drug interactions program

Alcohol (Ethyl): CNS Depressants may enhance the CNS depressant effect of Alcohol (Ethyl). Risk C: Monitor therapy

Alizapride: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Azelastine (Nasal): May enhance the CNS depressant effect of CNS Depressants. Risk X: Avoid combination

Blonanserin: CNS Depressants may enhance the CNS depressant effect of Blonanserin. Management: Use caution if coadministering blonanserin and CNS depressants; dose reduction of the other CNS depressant may be required. Strong CNS depressants should not be coadministered with blonanserin. Risk D: Consider therapy modification

Brexanolone: CNS Depressants may enhance the CNS depressant effect of Brexanolone. Risk C: Monitor therapy

Brimonidine (Topical): May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Bromopride: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Bromperidol: May enhance the CNS depressant effect of CNS Depressants. Risk X: Avoid combination

Buprenorphine: CNS Depressants may enhance the CNS depressant effect of Buprenorphine. Management: Consider reduced doses of other CNS depressants, and avoiding such drugs in patients at high risk of buprenorphine overuse/self-injection. Initiate buprenorphine at lower doses in patients already receiving CNS depressants. Risk D: Consider therapy modification

Cannabinoid-Containing Products: CNS Depressants may enhance the CNS depressant effect of Cannabinoid-Containing Products. Risk C: Monitor therapy

Chlormethiazole: May enhance the CNS depressant effect of CNS Depressants. Management: Monitor closely for evidence of excessive CNS depression. The chlormethiazole labeling states that an appropriately reduced dose should be used if such a combination must be used. Risk D: Consider therapy modification

Chlorphenesin Carbamate: May enhance the adverse/toxic effect of CNS Depressants. Risk C: Monitor therapy

CNS Depressants: May enhance the adverse/toxic effect of other CNS Depressants. Risk C: Monitor therapy

CYP2B6 Inducers (Moderate): May decrease the serum concentration of Ketamine. Risk C: Monitor therapy

CYP3A4 Inducers (Moderate): May decrease the serum concentration of Ketamine. Risk C: Monitor therapy

CYP3A4 Inducers (Strong): May decrease the serum concentration of Ketamine. Risk C: Monitor therapy

CYP3A4 Inhibitors (Strong): May increase the serum concentration of Ketamine. Risk C: Monitor therapy

Daridorexant: May enhance the CNS depressant effect of CNS Depressants. Management: Dose reduction of daridorexant and/or any other CNS depressant may be necessary. Use of daridorexant with alcohol is not recommended, and the use of daridorexant with any other drug to treat insomnia is not recommended. Risk D: Consider therapy modification

DexmedeTOMIDine: CNS Depressants may enhance the CNS depressant effect of DexmedeTOMIDine. Management: Monitor for increased CNS depression during coadministration of dexmedetomidine and CNS depressants, and consider dose reductions of either agent to avoid excessive CNS depression. Risk D: Consider therapy modification

Difelikefalin: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Dimethindene (Topical): May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Doxylamine: May enhance the CNS depressant effect of CNS Depressants. Management: The manufacturer of Diclegis (doxylamine/pyridoxine), intended for use in pregnancy, specifically states that use with other CNS depressants is not recommended. Risk C: Monitor therapy

Droperidol: May enhance the CNS depressant effect of CNS Depressants. Management: Consider dose reductions of droperidol or of other CNS agents (eg, opioids, barbiturates) with concomitant use. Risk D: Consider therapy modification

Esketamine: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Fexinidazole: May decrease the serum concentration of CYP2B6 Substrates (High risk with Inducers). Management: Avoid concomitant use of fexinidazole and CYP2B6 substrates when possible. If combined, monitor for reduced efficacy of the CYP2B6 substrate. Risk D: Consider therapy modification

Flunarizine: CNS Depressants may enhance the CNS depressant effect of Flunarizine. Risk X: Avoid combination

Flunitrazepam: CNS Depressants may enhance the CNS depressant effect of Flunitrazepam. Management: Reduce the dose of CNS depressants when combined with flunitrazepam and monitor patients for evidence of CNS depression (eg, sedation, respiratory depression). Use non-CNS depressant alternatives when available. Risk D: Consider therapy modification

Grapefruit Juice: May increase the serum concentration of Ketamine. Risk C: Monitor therapy

HydrOXYzine: May enhance the CNS depressant effect of CNS Depressants. Management: Consider a decrease in the CNS depressant dose, as appropriate, when used together with hydroxyzine. Increase monitoring of signs/symptoms of CNS depression in any patient receiving hydroxyzine together with another CNS depressant. Risk D: Consider therapy modification

Kava Kava: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Kratom: May enhance the CNS depressant effect of CNS Depressants. Risk X: Avoid combination

Lemborexant: May enhance the CNS depressant effect of CNS Depressants. Management: Dosage adjustments of lemborexant and of concomitant CNS depressants may be necessary when administered together because of potentially additive CNS depressant effects. Close monitoring for CNS depressant effects is necessary. Risk D: Consider therapy modification

Lisuride: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Lofexidine: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Lumacaftor and Ivacaftor: May decrease the serum concentration of CYP2B6 Substrates (High risk with Inducers). Risk C: Monitor therapy

Magnesium Sulfate: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Memantine: NMDA Receptor Antagonists may enhance the adverse/toxic effect of Memantine. Risk C: Monitor therapy

Methotrimeprazine: CNS Depressants may enhance the CNS depressant effect of Methotrimeprazine. Methotrimeprazine may enhance the CNS depressant effect of CNS Depressants. Management: Reduce the usual dose of CNS depressants by 50% if starting methotrimeprazine until the dose of methotrimeprazine is stable. Monitor patient closely for evidence of CNS depression. Risk D: Consider therapy modification

Metoclopramide: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

MetyroSINE: CNS Depressants may enhance the sedative effect of MetyroSINE. Risk C: Monitor therapy

MiFEPRIStone: May increase the serum concentration of CYP2B6 Substrates (High risk with Inhibitors). Risk C: Monitor therapy

Minocycline (Systemic): May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Mivacurium: Ketamine may enhance the therapeutic effect of Mivacurium. Risk C: Monitor therapy

Olopatadine (Nasal): May enhance the CNS depressant effect of CNS Depressants. Risk X: Avoid combination

Opioid Agonists: CNS Depressants may enhance the CNS depressant effect of Opioid Agonists. Management: Avoid concomitant use of opioid agonists and benzodiazepines or other CNS depressants when possible. These agents should only be combined if alternative treatment options are inadequate. If combined, limit the dosages and duration of each drug. Risk D: Consider therapy modification

Orphenadrine: CNS Depressants may enhance the CNS depressant effect of Orphenadrine. Risk X: Avoid combination

Oxomemazine: May enhance the CNS depressant effect of CNS Depressants. Risk X: Avoid combination

Oxybate Salt Products: CNS Depressants may enhance the CNS depressant effect of Oxybate Salt Products. Management: Consider alternatives to this combination when possible. If combined, dose reduction or discontinuation of one or more CNS depressants (including the oxybate salt product) should be considered. Interrupt oxybate salt treatment during short-term opioid use Risk D: Consider therapy modification

OxyCODONE: CNS Depressants may enhance the CNS depressant effect of OxyCODONE. Management: Avoid concomitant use of oxycodone and benzodiazepines or other CNS depressants when possible. These agents should only be combined if alternative treatment options are inadequate. If combined, limit the dosages and duration of each drug. Risk D: Consider therapy modification

Paraldehyde: CNS Depressants may enhance the CNS depressant effect of Paraldehyde. Risk X: Avoid combination

Perampanel: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Piribedil: CNS Depressants may enhance the CNS depressant effect of Piribedil. Risk C: Monitor therapy

Pramipexole: CNS Depressants may enhance the sedative effect of Pramipexole. Risk C: Monitor therapy

Procarbazine: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Ropeginterferon Alfa-2b: CNS Depressants may enhance the adverse/toxic effect of Ropeginterferon Alfa-2b. Specifically, the risk of neuropsychiatric adverse effects may be increased. Management: Avoid coadministration of ropeginterferon alfa-2b and other CNS depressants. If this combination cannot be avoided, monitor patients for neuropsychiatric adverse effects (eg, depression, suicidal ideation, aggression, mania). Risk D: Consider therapy modification

ROPINIRole: CNS Depressants may enhance the sedative effect of ROPINIRole. Risk C: Monitor therapy

Rotigotine: CNS Depressants may enhance the sedative effect of Rotigotine. Risk C: Monitor therapy

Rufinamide: May enhance the adverse/toxic effect of CNS Depressants. Specifically, sleepiness and dizziness may be enhanced. Risk C: Monitor therapy

Suvorexant: CNS Depressants may enhance the CNS depressant effect of Suvorexant. Management: Dose reduction of suvorexant and/or any other CNS depressant may be necessary. Use of suvorexant with alcohol is not recommended, and the use of suvorexant with any other drug to treat insomnia is not recommended. Risk D: Consider therapy modification

Thalidomide: CNS Depressants may enhance the CNS depressant effect of Thalidomide. Risk X: Avoid combination

Theophylline Derivatives: Ketamine may enhance the adverse/toxic effect of Theophylline Derivatives. Specifically, the risk for seizures may be increased. Risk C: Monitor therapy

Thiopental: Ketamine may enhance the adverse/toxic effect of Thiopental. Risk C: Monitor therapy

Thiotepa: May increase the serum concentration of CYP2B6 Substrates (High risk with Inhibitors). Risk C: Monitor therapy

Ticlopidine: May increase the serum concentration of Ketamine. Risk C: Monitor therapy

Trimeprazine: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Valerian: May enhance the CNS depressant effect of CNS Depressants. Risk C: Monitor therapy

Zolpidem: CNS Depressants may enhance the CNS depressant effect of Zolpidem. Management: Reduce the Intermezzo brand sublingual zolpidem adult dose to 1.75 mg for men who are also receiving other CNS depressants. No such dose change is recommended for women. Avoid use with other CNS depressants at bedtime; avoid use with alcohol. Risk D: Consider therapy modification

Pregnancy Considerations

Ketamine crosses the placenta (Ellingson 1977; Little 1972).

Ketamine produces dose dependent increases in uterine contractions; effects may vary by trimester. The plasma clearance of ketamine is reduced during pregnancy. Dose related neonatal depression and decreased Apgar scores have been reported with large doses administered at delivery (Little 1972; Neuman 2013; White 1982).

Based on animal data, repeated or prolonged use of general anesthetic and sedation medications that block N-methyl-D-aspartate (NMDA) receptors and/or potentiate gamma-aminobutyric acid (GABA) activity may affect brain development. Evaluate benefits and potential risks of fetal exposure to ketamine when duration of surgery is expected to be >3 hours (Olutoye 2018).

Although obstetric use is not recommended by the manufacturer, ketamine has been evaluated for use during cesarean and vaginal delivery (ACOG 209 2019; Akamatsu 1974; Galbert 1973). Ketamine may be considered as an alternative induction agent in females requiring general anesthesia for cesarean delivery who are hemodynamically unstable (Devroe 2015). Use of ketamine as an adjunctive analgesic in cesarean section has also been evaluated; however, use for this purpose may require additional studies (Carvalho 2017; Heesen 2015). When sedation and analgesia is needed for other procedures during pregnancy, low doses of ketamine may be used, but other agents are preferred (Neuman 2013; Schwenk 2018). Use of ketamine infusion for the treatment of refractory status epilepticus in a pregnant patient has been noted in a case report (Talahma 2018).

The ACOG recommends that pregnant women should not be denied medically necessary surgery, regardless of trimester. If the procedure is elective, it should be delayed until after delivery (ACOG 775 2019).

Monitoring Parameters

Heart rate, BP, respiratory rate, transcutaneous O2 saturation, emergence reactions; cardiac function (continuously monitored in older patients with increased BP or cardiac decompensation); LFTs, alkaline phosphatase, and gamma glutamyl transferase (baseline and then at periodic intervals).

Mechanism of Action

Produces a cataleptic-like state in which the patient is dissociated from the surrounding environment by direct action on the cortex and limbic system. Ketamine is a noncompetitive NMDA receptor antagonist that blocks glutamate. Low (subanesthetic) doses produce analgesia, and modulate central sensitization, hyperalgesia and opioid tolerance. Reduces polysynaptic spinal reflexes.

Pharmacokinetics (Adult data unless noted)

Onset of action:

IV: Anesthetic effect: Within 30 seconds

IM: Anesthetic effect: 3 to 4 minutes; Analgesia: Within 10 to 15 minutes

Intranasal: Analgesic effect: Within 10 minutes (Carr 2004); Sedation: Children 2 to 6 years: 5 to 8 minutes (Bahetwar 2011)

Oral: Analgesia: Within 30 minutes; Sedation: Children 2 to 8 years (Turhanoglu 2003):

4 mg/kg/dose: 12.9 ± 1.9 minutes

6 mg/kg/dose: 10.4 ± 2.9 minutes

8 mg/kg/dose: 9.5 ± 1.9 minutes

Duration:

IV: Anesthetic effect: 5 to 10 minutes; Recovery: 1 to 2 hours

IM: Anesthetic effect: 12 to 25 minutes; Analgesia: 15 to 30 minutes; Recovery: 3 to 4 hours

Intranasal: Analgesic effect: Up to 60 minutes (Carr 2004); Recovery: Children 2 to 6 years: 34 to 46 minutes (Bahetwar 2011)

Distribution: Vdss: 2.1 to 3.1 L/kg (Clements 1981)

Protein binding: 27% (Brunton 2006)

Metabolism: Hepatic via N-dealkylation (metabolite I [norketamine]), hydroxylation of the cyclohexone ring (metabolites III and IV), conjugation with glucuronic acid and dehydration of the hydroxylated metabolites to form the cyclohexene derivative (metabolite II); metabolite I (norketamine) is 33% as potent as parent compound. When administered orally, norketamine concentrations are higher compared to other routes of administration due to extensive first-pass metabolism in the liver (Blonk 2010; Soto 2012).

Bioavailability:

IM: 93%

Oral: 20% to 30% (Miller 2010)

Intranasal: Children, Adolescents, and Adults: Mean range: 35% to 50% (Malinovsky 1996; Miller 2010; Nielsen 2014)

Rectal: Children 2 to 9 years: 25% (Malinovsky 1996)

Half-life elimination: Alpha: 10 to 15 minutes; Beta: 2.5 hours

Time to peak, plasma:

IM: 5 to 30 minutes (Clements 1982)

Intranasal: 10 to 14 minutes (Huge 2010); Children 2 to 9 years: ~20 minutes (Malinovsky 1996)

Oral: ~30 minutes (Soto 2012)

Rectal: Children 2 to 9 years: ~45 minutes (Malinovsky 1996)

Excretion: Urine (91%); feces (3%) (Ghoneim 1977)

Pricing: US

Solution (Ketalar Injection)

10 mg/mL (per mL): $1.19

50 mg/mL (per mL): $0.84

100 mg/mL (per mL): $3.20

Solution (Ketamine HCl Injection)

10 mg/mL (per mL): $1.21

50 mg/mL (per mL): $0.43 - $0.98

100 mg/mL (per mL): $1.99 - $3.06

Disclaimer: A representative AWP (Average Wholesale Price) price or price range is provided as reference price only. A range is provided when more than one manufacturer's AWP price is available and uses the low and high price reported by the manufacturers to determine the range. The pricing data should be used for benchmarking purposes only, and as such should not be used alone to set or adjudicate any prices for reimbursement or purchasing functions or considered to be an exact price for a single product and/or manufacturer. Medi-Span expressly disclaims all warranties of any kind or nature, whether express or implied, and assumes no liability with respect to accuracy of price or price range data published in its solutions. In no event shall Medi-Span be liable for special, indirect, incidental, or consequential damages arising from use of price or price range data. Pricing data is updated monthly.

Brand Names: International
  • Aneket (ET, LK);
  • Callypsol (TH);
  • Calypsol (AE, BB, BG, BM, BS, BZ, CY, CZ, GY, HN, HU, IQ, IR, JM, JO, LB, LY, OM, PK, PL, PR, RO, SA, SR, SY, TT, VN, YE);
  • Clortamina (BR);
  • Cost (AR);
  • Dorgipain (EG);
  • Etamine (PH);
  • Kain (BD);
  • Kanox (MY);
  • Keiran (VE);
  • Ketalar (AE, AU, BB, BD, BE, BM, BR, BS, BZ, CH, CY, DK, EE, FI, FR, GB, GR, GY, HK, IE, IL, IN, IQ, IR, IT, JM, JO, LB, LU, LY, MT, MY, NO, NZ, OM, PE, PT, SA, SE, SK, SR, SY, TH, TR, TT, UY, YE);
  • Ketalin (MX);
  • Ketam (BD, EG, ET);
  • Ketamar (EG);
  • Ketamax (PH);
  • Ketamin-S (+) (PY);
  • Ketamina (CU);
  • Ketamine-P (TH);
  • Ketanest (AR, AT, HR, PL);
  • Ketanir (LK);
  • Ketaride (BD);
  • Ketashort (CO);
  • Ketava (MY);
  • Ketavit (LK);
  • Ketmin (IN);
  • Ketolar (ES);
  • Ketomin (KR);
  • Narkamon (DE, PL);
  • Raketiv (BE);
  • Spravato (HR);
  • Tekam (AE, BH, CY, EG, IQ, IR, JO, LB, LY, OM, QA, SA, SY, YE);
  • Velonarcon (PL)


For country code abbreviations (show table)
  1. aan het Rot M, Collins KA, Murrough JW, et al. Safety and efficacy of repeated-dose intravenous ketamine for treatment-resistant depression. Biol Psychiatry. 2010;67(2):139-145. doi:10.1016/j.biopsych.2009.08.038 [PubMed 19897179]
  2. Adhikary SD, Thiruvenkatarajan V, McFadden A, Liu WM, Mets B, Rogers A. Analgesic efficacy of ketamine and magnesium after laparoscopic sleeve gastrectomy: a randomized, double-blind, placebo-controlled trial. J Clin Anesth. 2021;68:110097. doi:10.1016/j.jclinane.2020.110097 [PubMed 33120301]
  3. Akamatsu TJ, Bonica JJ, Rehmet R, Eng M, Ueland K. Experiences with the use of ketamine for parturition. I. Primary anesthetic for vaginal delivery. Anesth Analg. 1974;53(2):284-287. [PubMed 4856139]
  4. Albanèse J, Arnaud S, Rey M, Thomachot L, Alliez B, Martin C. Ketamine decreases intracranial pressure and electroencephalographic activity in traumatic brain injury patients during propofol sedation. Anesthesiology. 1997;87(6):1328-1334. doi:10.1097/00000542-199712000-00011 [PubMed 9416717]
  5. Alletag MJ, Auerbach MA, Baum CR. Ketamine, propofol, and ketofol use for pediatric sedation. Pediatr Emerg Care. 2012;28(12):1391-1395; quiz 1396-8. doi:10.1097/PEC.0b013e318276fde2. [PubMed 23222112]
  6. American College of Obstetricians and Gynecologists (ACOG). ACOG committee opinion no. 775: nonobstetric surgery during pregnancy. Obstet Gynecol. 2019;133(4):e285-e286. doi:10.1097/AOG.0000000000003174 [PubMed 30913200]
  7. American College of Obstetricians and Gynecologists (ACOG). ACOG practice bulletin no. 209: obstetric analgesia and anesthesia. Obstet Gynecol. 2019;133(3):e208-e225. doi:10.1097/AOG.0000000000003132 [PubMed 30801474]
  8. Anand KJ, International Evidence-Based Group for Neonatal Pain. Consensus statement for the prevention and management of pain in the newborn. Arch Pediatr Adolesc Med. 2001;155(2):173-180. doi:10.1001/archpedi.155.2.173 [PubMed 11177093]
  9. 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(6):504-512.e1-e2. doi:10.1016/j.annemergmed.2012.01.017 [PubMed 22401952]
  10. Andolfatto G, Willman E, Joo D, et al. Intranasal ketamine for analgesia in the emergency department: a prospective observational series. Acad Emerg Med. 2013;20(10):1050-1054. doi:10.1111/acem.12229 [PubMed 24127709]
  11. Aroni F, Iacovidou N, Dontas I, Pourzitaki C, Xanthos T. Pharmacological aspects and potential new clinical applications of ketamine: reevaluation of an old drug. J Clin Pharmacol. 2009;49(8):957-964. doi:10.1177/0091270009337941 [PubMed 19546251]
  12. Asadi P, Ghafouri HB, Yasinzadeh M, Kasnavieh SM, Modirian E. Ketamine and atropine for pediatric sedation: a prospective double-blind randomized controlled trial. Pediatr Emerg Care. 2013;29(2):136-139. doi:10.1097/PEC.0b013e31828058b2. [PubMed 23364373]
  13. Bahetwar SK, Pandey RK, Saksena AK, Chandra G. A comparative evaluation of intranasal midazolam, ketamine and their combination for sedation of young uncooperative pediatric dental patients: a triple blind randomized crossover trial. J Clin Pediatr Dent. 2011;35(4):415-420. doi:10.17796/jcpd.35.4.l43h3354705u2574 [PubMed 22046702]
  14. Ballow SL, Kaups KL, Anderson S, Chang M. A standardized rapid sequence intubation protocol facilitates airway management in critically injured patients. J Trauma Acute Care Surg. 2012;73(6):1401-1405. doi:10.1097/TA.0b013e318270dcf5. [PubMed 23188232]
  15. Barkan S, Breitbart R, Brenner-Zada G, et al. A double-blind, randomised, placebo-controlled trial of oral midazolam plus oral ketamine for sedation of children during laceration repair. Emerg Med J. 2014;31(8):649-653. doi:10.1136/emermed-2012-202189. [PubMed 23686730]
  16. Barnard EB, Moy RJ, Kehoe AD, Bebarta VS, Smith JE. Rapid sequence induction of anaesthesia via the intraosseous route: a prospective observational study. Emerg Med J. 2015;32(6):449-452. doi:10.1136/emermed-2014-203740 [PubMed 24963149]
  17. Barois J, Tourneux P. Ketamine and atropine decrease pain for preterm newborn tracheal intubation in the delivery room: an observational pilot study. Acta Paediatr. 2013;102(12):e534-538. doi:10.1111/apa.12413 [PubMed 24015945]
  18. Barr J, Fraser GL, Puntillo K, et al. Clinical practice guidelines for the management of pain, agitation, and delirium in adult patients in the intensive care unit. Crit Care Med. 2013;41(1):263-306. doi:10.1097/CCM.0b013e3182783b72 [PubMed 23269131]
  19. Based on expert opinion.
  20. Beaudequin D, Can AT, Dutton M, et al. Predicting therapeutic response to oral ketamine for chronic suicidal ideation: a Bayesian network for clinical decision support. BMC Psychiatry. 2020;20(1):519. doi:10.1186/s12888-020-02925-1 [PubMed 33115424]
  21. Beebe DS, Belani KG, Chang PN, et al. Effectiveness of preoperative sedation with rectal midazolam, ketamine, or their combination in young children. Anesth Analg. 1992;75(6):880-884. doi:10.1213/00000539-199212000-00003 [PubMed 1443705]
  22. Berkenbosch JW, Graff GR, Stark JM. Safety and efficacy of ketamine sedation for infant flexible fiberoptic bronchoscopy. Chest. 2004;125(3):1132-1137. doi:10.1378/chest.125.3.1132 [PubMed 15006978]
  23. Blonk MI, Koder BG, van den Bemt PM, Huygen FJ. Use of oral ketamine in chronic pain management: a review. Eur J Pain. 2010;14(5):466-472. doi:10.1016/j.ejpain.2009.09.005 [PubMed 19879174]
  24. Bovill JG. Intravenous anesthesia for the patient with left ventricular dysfunction. Semin Cardiothorac Vasc Anesth. 2006 Mar;10(1):43-8. doi: 10.1177/108925320601000108. PMID: 16703233. [PubMed 16703233]
  25. Bowles ED, Gold ME. Rethinking the paradigm: evaluation of ketamine as a neurosurgical anesthetic [published correction appears in AANA J. 2013;81(1):18.]. AANA J. 2012;80(6):445-452. [PubMed 23409639]
  26. Bravenec B. General anesthesia: Intravenous induction agents. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. Accessed July 29, 2020. http://www.uptodate.com.
  27. Brophy GM, Bell R, Claassen J, et al; Neurocritical Care Society Status Epilepticus Guideline Writing Committee. Guidelines for the evaluation and management of status epilepticus. Neurocrit Care. 2012;17(1):3-23. doi:10.1007/s12028-012-9695-z [PubMed 22528274]
  28. Brown CA III, Sakles JC. Rapid sequence intubation for adults outside the operating room. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. Accessed July 29, 2020. http://www.uptodate.com.
  29. Brunton LL, Lazo JS, Park KL, eds. Goodman & Gilman's The Pharmacological Basis of Therapeutics. 11th ed. New York, NY: McGraw-Hill Medical; 2006.
  30. Buchheit JL, Yeh DD, Eikermann M, Lin H. Impact of low-dose ketamine on the usage of continuous opioid infusion for the treatment of pain in adult mechanically ventilated patients in surgical intensive care units. J Intensive Care Med. 2019;34(8):646-651. doi:10.1177/0885066617706907 [PubMed 28468568]
  31. Cabbabe EB, Behbahani PM. Cardiovascular reactions associated with the use of ketamine and epinephrine in plastic surgery. Ann Plast Surg. 1985;15(1):50-56. doi:10.1097/00000637-198507000-00006 [PubMed 2417535]
  32. Caro D. Induction agents for rapid sequence intubation in adults outside the operating room. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. Accessed July 29, 2020. http://www.uptodate.com.
  33. Carr DB, Goudas LC, Denman WT, et al. Safety and efficacy of intranasal ketamine for the treatment of breakthrough pain in patients with chronic pain: a randomized, double-blind, placebo-controlled, crossover study. Pain. 2004;108(1-2):17-27. doi:10.1016/j.pain.2003.07.001 [PubMed 15109503]
  34. Carvalho B, Butwick AJ. Postcesarean delivery analgesia. Best Pract Res Clin Anaesthesiol. 2017;31(1):69-79. doi:10.1016/j.bpa.2017.01.003. [PubMed 28625307]
  35. Castilla M, Jerez M, Llácer M, Martinez S. Anaesthetic management in a neonate with congenital complete heart block. Paediatr Anaesth. 2004;14(2):172-175. doi:10.1046/j.1460-9592.2003.01180.x [PubMed 14962334]
  36. Chen CH, Lee MH, Chen YC, Lin MF. Ketamine-snorting associated cystitis. J Formos Med Assoc. 2011;110(12):787-791. doi:10.1016/j.jfma.2011.11.010 [PubMed 22248834]
  37. Cherng CH, Wong CS. Ketamine-induced emergence reactions after desflurane anaesthesia. Eur J Anaesthesiol. 2007;24(2):200-201. doi:10.1017/S0265021506001773 [PubMed 17038218]
  38. Chow TG, McConnell J, Lee MJ. A 2-year-old girl with periprocedural anaphylaxis. Allergy Asthma Proc. 2021;42(1):97-99. doi:10.2500/aap.2021.42.200031 [PubMed 33404393]
  39. 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(3):228-235. doi:10.1111/j.1553-2712.2000.tb01064.x [PubMed 10730829]
  40. Clements JA, Nimmo WS, Grant IS. Bioavailability, pharmacokinetics, and analgesic activity of ketamine in humans. J Pharm Sci. 1982;71(5):539-542. doi:10.1002/jps.2600710516 [PubMed 7097501]
  41. Clements JA, Nimmo WS. Pharmacokinetics and analgesic effect of ketamine in man. Br J Anaesth. 1981;53(1):27-30. doi:10.1093/bja/53.1.27 [PubMed 7459184]
  42. Cohen L, Athaide V, Wickham ME, Doyle-Waters MM, Rose NG, Hohl CM. The effect of ketamine on intracranial and cerebral perfusion pressure and health outcomes: a systematic review. Ann Emerg Med. 2015;65(1):43-51.e2. doi:10.1016/j.annemergmed.2014.06.018 [PubMed 25064742]
  43. Cohen SP, Bhatia A, Buvanendran A, et al. Consensus guidelines on the use of intravenous ketamine infusions for chronic pain from the American Society of Regional Anesthesia and Pain Medicine, the American Academy of Pain Medicine, and the American Society of Anesthesiologists. Reg Anesth Pain Med. 2018;43(5):521-546. doi:10.1097/AAP.0000000000000808 [PubMed 29870458]
  44. Cole JB, Klein LR, Nystrom PC, et al. A prospective study of ketamine as primary therapy for prehospital profound agitation. Am J Emerg Med. 2018;36(5):789-796. doi:10.1016/j.ajem.2017.10.022 [PubMed 29033344]
  45. Cole JB, Moore JC, Nystrom PC, et al. A prospective study of ketamine versus haloperidol for severe prehospital agitation. Clin Toxicol (Phila). 2016;54(7):556-562. doi:10.1080/15563650.2016.1177652. [PubMed 27102743]
  46. Corrigan M, Wilson SS, Hampton J. Safety and efficacy of intranasally administered medications in the emergency department and prehospital settings. Am J Health Syst Pharm. 2015;72(18):1544-1554. doi:10.2146/ajhp140630 [PubMed 26346210]
  47. Cosci F, Chouinard G. Acute and persistent withdrawal syndromes following discontinuation of psychotropic medications. Psychother Psychosom. 2020;89(5):283-306. doi:10.1159/000506868 [PubMed 32259826]
  48. Cote CJ. Sedation for the Pediatric Patient: A Review. Pediatr Clin North Am. 1994;41(1):31-58. doi:10.1016/s0031-3955(16)38690-4 [PubMed 8295806]
  49. Coté CJ, Lerman J, Anderson BJ, eds. A Practice of Anesthesia for Infants and Children. 5th ed. Philadelphia, PA: Elsevier Saunders; 2013.
  50. Coté CJ, Lerman J, Anderson B, eds. A Practice of Anesthesia for Infants and Children. 6th ed. Elsevier; 2019.
  51. Cotter S, Wong J, Gada N, et al. Repeated or continuous medically supervised ketamine administration associated with hepatobiliary adverse events: a retrospective case series. Drug Saf. 2021;44(12):1365-1374. doi:10.1007/s40264-021-01120-9 [PubMed 34699023]
  52. Critchlow DG. A case of ketamine dependence with discontinuation symptoms. Addiction. 2006;101(8):1212-1213. doi:10.1111/j.1360-0443.2006.01494.x [PubMed 16869851]
  53. Cunningham AJ, Barry P. Intraocular pressure--physiology and implications for anaesthetic management. Can Anaesth Soc J. 1986 Mar;33(2):195-208. doi:10.1007/BF03010831 [PubMed 3516335]
  54. David H, Shipp J. A randomized controlled trial of ketamine/propofol versus propofol alone for emergency department procedural sedation. Ann Emerg Med. 2011;57(5):435-441. doi:10.1016/j.annemergmed.2010.11.025 [PubMed 21256626]
  55. Davis J, Bates L. Rapid sequence induction via an intraosseous needle. J Intensive Care Soc. 2016;17(2):178-179. doi:10.1177/1751143715614845 [PubMed 28979485]
  56. Denmark TK, Crane HA, Brown L. Ketamine to avoid mechanical ventilation in severe pediatric asthma. J Emerg Med. 2006;30(2):163-166. doi:10.1016/j.jemermed.2005.09.003 [PubMed 16567251]
  57. Devlin JW, Skrobik Y, Gélinas C, et al; Society of Critical Care Medicine. Clinical practice guidelines for the prevention and management of pain, agitation/sedation, delirium, immobility, and sleep disruption in adult patients in the ICU. Crit Care Med. 2018;46(9):e825-e873. doi:10.1097/CCM.0000000000003299 [PubMed 30113379]
  58. Devroe S, Van de Velde M, Rex S. General anesthesia for caesarean section. Curr Opin Anaesthesiol. 2015;28(3):240-246. doi:10.1097/ACO.0000000000000185. [PubMed 25827280]
  59. Diaz JH. Intranasal ketamine preinduction of paediatric outpatients. Paediatr Anaesth. 1997;7(4):273-278. doi:10.1046/j.1460-9592.1997.d01-93.x [PubMed 9243683]
  60. Drayna PC, Estrada C, Wang W, Saville BR, Arnold DH. Ketamine sedation is not associated with clinically meaningful elevation of intraocular pressure. Am J Emerg Med. 2012;30(7):1215-1218. doi:10.1016/j.ajem.2011.06.001. [PubMed 22169582]
  61. Driver BE, Reardon RF. Apnea after low-dose ketamine sedation during attempted delayed sequence intubation. Ann Emerg Med. 2017;69(1):34-35. doi:10.1016/j.annemergmed.2016.07.026 [PubMed 27692755]
  62. Ellingson A, Haram K, Sagen N, Solheim E. Transplacental passage of ketamine after intravenous administration. Acta Anaesthesiol Scand. 1977;21(1):41-44. doi:10.1111/j.1399-6576.1977.tb01191.x [PubMed 842268]
  63. Erstad BL, Barletta JF. Drug dosing in the critically ill obese patient-a focus on sedation, analgesia, and delirium. Crit Care. 2020;24(1):315. doi:10.1186/s13054-020-03040-z [PubMed 32513237]
  64. Fantacci C, Fabrizio GC, Ferrara P, Franceschi F, Chiaretti A. Intranasal drug administration for procedural sedation in children admitted to pediatric emergency room. Eur Rev Med Pharmacol Sci. 2018;22(1):217-222. doi:10.26355/eurrev_201801_14120 [PubMed 29364490]
  65. Fava M, Freeman MP, Flynn M, et al. Double-blind, placebo-controlled, dose-ranging trial of intravenous ketamine as adjunctive therapy in treatment-resistant depression (TRD). Mol Psychiatry. 2020;25(7):1592-1603. doi:10.1038/s41380-018-0256-5 [PubMed 30283029]
  66. FDA Drug Safety Communication. FDA review results in new warnings about using general anesthetics and sedation drugs in young children and pregnant women. Food and Drug Administration website. Accessed May 26, 2017. https://www.fda.gov/Drugs/DrugSafety/ucm532356.htm.
  67. Ferguson CL, Beckett RD. Intranasal ketamine for treatment of acute pain in pediatrics: a systematic review. Pediatr Emerg Care. 2020;36(8):e476-e481. doi:10.1097/PEC.0000000000002181 [PubMed 32665505]
  68. Fishman DS, Andropoulos DB, Lightdale JR. Sedation and the Food and Drug Administration warning: what a pediatric gastroenterologist, hepatologist, and pancreatologist should know. J Pediatr Gastroenterol Nutr. 2019;69(1):3-5. doi:10.1097/MPG.0000000000002346 [PubMed 30921252]
  69. Fond G, Loundou A, Rabu C, et al. Ketamine administration in depressive disorders: a systematic review and meta-analysis. Psychopharmacology (Berl). 2014;231(18):3663-3676. doi:10.1007/s00213-014-3664-5 [PubMed 25038867]
  70. Frey TM, Florin TA, Caruso M, Zhang N, Zhang Y, Mittiga MR. Effect of intranasal ketamine vs fentanyl on pain reduction for extremity injuries in children: the PRIME randomized clinical trial. JAMA Pediatr. 2019;173(2):140-146. doi:10.1001/jamapediatrics.2018.4582 [PubMed 30592476]
  71. Fuhrman B, Zimmerman J, eds. Pediatric Critical Care. 4th ed. Philadelphia, PA: Elsevier Health; 2011.
  72. Galbert MW, Gardner AE. Ketamine for obstetrical anesthesia. Anesth Analg. 1973;52(6):926-930. [PubMed 4796568]
  73. Garber PM, Droege CA, Carter KE, Harger NJ, Mueller EW. Continuous infusion ketamine for adjunctive analgosedation in mechanically ventilated, critically ill patients. Pharmacotherapy. 2019;39(3):288-296. doi:10.1002/phar.2223 [PubMed 30746728]
  74. Garg A, Sinha P, Kumar P, Prakash O. Use of naltrexone in ketamine dependence. Addict Behav. 2014;39(8):1215-1216. doi:10.1016/j.addbeh.2014.04.004 [PubMed 24813545]
  75. Gaspard N, Foreman B, Judd LM, et al. Intravenous ketamine for the treatment of refractory status epilepticus: a retrospective multicenter study. Epilepsia. 2013;54(8):1498-1503. doi:10.1111/epi.12247 [PubMed 23758557]
  76. Gautam SN, Bhatta S, Sangraula D, Shrestha BC, Rawal SB. Intranasal midazolam vs ketamine as premedication in paediatric surgical procedure for child separation and induction. Nepal Med Coll J. 2007;9(3):179-181. [PubMed 18092436]
  77. Geiger-Hayes J. PAINWeek journal vol 6, Q4. Published 2018. Accessed July 29, 2020. https://www.painweek.org/media/journal/pwj-painweek-journal-vol-6-q4.
  78. Gelissen HP, Epema AH, Henning RH, Krijnen HJ, Hennis PJ, den Hertog A. Inotropic effects of propofol, thiopental, midazolam, etomidate, and ketamine on isolated human atrial muscle. Anesthesiology. 1996;84(2):397-403. doi:10.1097/00000542-199602000-00019 [PubMed 8602672]
  79. Ghoneim MM, Korttila K. Pharmacokinetics of Intravenous Anaesthetics: Implications for Clinical Use. Clin Pharmacokinet. 1977;2(5):344-372. doi:10.2165/00003088-197702050-00003 [PubMed 21053]
  80. Godwin SA, Burton JH, Gerardo CJ, et al; American College of Emergency Physicians. Clinical policy: procedural sedation and analgesia in the emergency department. Ann Emerg Med. 2014;63(2):247-258.e18. doi:10.1016/j.annemergmed.2013.10.015 [PubMed 24438649]
  81. Gómez-Revuelta M, Fernández-Rodríguez M, Boada-Antón L, Vázquez-Bourgon J. Apnea during slow sub-anaesthetic infusion of intravenous ketamine for treatment-resistant depression. Ther Adv Psychopharmacol. 2020;10:2045125320981498. doi:10.1177/2045125320981498 [PubMed 33489089]
  82. Goyal S, Ambekar A, Ray R. Ketamine dependence in an anesthesiologist: an occupational hazard? Indian J Psychol Med. 2014;36(3):335-337. doi:10.4103/0253-7176.135395 [PubMed 25035566]
  83. Grabowski J, Goldin A, Arthur LG, et al. The effects of early anesthesia on neurodevelopment: a systematic review. J Pediatr Surg. 2021:S0022-3468(21)00014-2. doi:10.1016/j.jpedsurg.2021.01.002 [PubMed 33509654]
  84. Graham MR. Clinical update regarding general anesthesia-associated neurotoxicity in infants and children. Curr Opin Anaesthesiol. 2017;30(6):682-687. doi:10.1097/ACO.0000000000000520 [PubMed 28915132]
  85. Graudins A, Meek R, Egerton-Warburton D, Oakley E, Seith R. The PICHFORK (Pain in Children Fentanyl or Ketamine) trial: a randomized controlled trial comparing intranasal ketamine and fentanyl for the relief of moderate to severe pain in children with limb injuries. Ann Emerg Med. 2015;65(3):248-254.e1. doi:10.1016/j.annemergmed.2014.09.024 [PubMed 25447557]
  86. Green SM, Roback MG, Kennedy RM, Krauss B. Clinical practice guideline for emergency department ketamine dissociative sedation: 2011 update. Ann Emerg Med. 2011;57(5):449-461. doi:10.1016/j.annemergmed.2010.11.030 [PubMed 21256625]
  87. Green SM, Roback MG, Krauss B, et al; Emergency Department Ketamine Meta-Analysis Study Group. Predictors of airway and respiratory adverse events with ketamine sedation in the emergency department: an individual-patient data meta-analysis of 8,282 children. Ann Emerg Med. 2009;54(2):158-68.e1-4. doi:10.1016/j.annemergmed.2008.12.011 [PubMed 19201064]
  88. Green SM, Sherwin TS. Incidence and severity of recovery agitation after ketamine sedation in young adults. Am J Emerg Med. 2005;23(2):142-144. doi:10.1016/j.ajem.2004.04.030 [PubMed 15765332]
  89. Groetzinger LM, Rivosecchi RM, Bain W, et al. Ketamine infusion for adjunct sedation in mechanically ventilated adults. Pharmacotherapy. 2018;38(2):181-188. doi:10.1002/phar.2065 [PubMed 29193185]
  90. Gropper MA, Eriksson LI, Fleisher LA, Wiener-Kronish JP, Cohen NH, Leslie K. Miller’s Anesthesia. 9th ed. Elsevier; 2019.
  91. Guerra GG, Robertson CM, Alton GY, et al; Western Canadian Complex Pediatric Therapies Follow-up Group. Neurodevelopmental outcome following exposure to sedative and analgesic drugs for complex cardiac surgery in infancy. Paediatr Anaesth. 2011;21(9):932-941. doi:10.1111/j.1460-9592.2011.03581.x [PubMed 21507125]
  92. Guthrie AM, Baum RA, Carter C, et al. Use of intranasal ketamine in pediatric patients in the emergency department. Pediatr Emerg Care. Published online July 8, 2019. doi:10.1097/PEC.0000000000001863 [PubMed 31290798]
  93. Gutstein HB, Johnson KL, Heard MB, Gregory GA. Oral ketamine preanesthetic medication in children. Anesthesiology. 1992;76(1):28-33. doi:10.1097/00000542-199201000-00004 [PubMed 1729932]
  94. Haas DA, Harper DG. Ketamine: a review of its pharmacologic properties and use in ambulatory anesthesia. Anesth Prog. 1992;39(3):61-68. [PubMed 1308374]
  95. Hall RW. Anesthesia and analgesia in the NICU. Clin Perinatol. 2012;39(1):239-254. [PubMed 22341549]
  96. Hatab SZ, Singh A, Felner EI, Kamat P. Transient central diabetes insipidus induced by ketamine infusion. Ann Pharmacother. 2014;48(12):1642-1645. doi:10.1177/1060028014549991 [PubMed 25225198]
  97. Heesen M, Böhmer J, Brinck EC, Kontinen VK, Klöhr S, Rossaint R, Straube S. Intravenous ketamine during spinal and general anaesthesia for caesarean section: systematic review and meta-analysis. Acta Anaesthesiol Scand. 2015;59(4):414-426. doi:10.1111/aas.12468. [PubMed 25789942]
  98. Hegenbarth MA; American Academy of Pediatrics Committee on Drugs. Preparing for pediatric emergencies: drugs to consider. Pediatrics. 2008;121(2):433-443.doi:10.1542/peds.2007-3284. [PubMed 18245435]
  99. Heinrich M, Wetzstein V, Muensterer OJ, Till H. Conscious sedation: Off-label use of rectal S(+)-ketamine and midazolam for wound dressing changes in paediatric heat injuries. Eur J Pediatr Surg. 2004;14(4):235-239. doi:10.1055/s-2004-817960 [PubMed 15343462]
  100. Heydari F, Gholamian A, Zamani M, Majidinejad S. Effect of intramuscular ketamine versus haloperidol on short-term control of severe agitated patients in emergency department; a randomized clinical trial. Bull Emerg Trauma. 2018;6(4):292-299. doi:10.29252/beat-060404 [PubMed 30402516]
  101. Hocking G, Cousins MJ. Ketamine in chronic pain management: an evidence-based review. Anesth Analg. 2003;97(6):1730-1739. doi:10.1213/01.ANE.0000086618.28845.9B [PubMed 14633551]
  102. Huge V, Lauchart M, Magerl W, et al. Effects of low-dose intranasal (S)-ketamine in patients with neuropathic pain. Eur J Pain. 2010;14(4):387-394. doi:10.1016/j.ejpain.2009.08.002 [PubMed 19733106]
  103. Ikonomidou C, Bosch F, Miksa M, et al. Blockade of NMDA receptors and apoptotic neurodegeneration in the developing brain. Science. 1999;283(5398):70-4. doi:10.1126/science.283.5398.70 [PubMed 9872743]
  104. Iqbal O'Meara AM, Miller Ferguson N, Zven SE, et al. Potential neurodevelopmental effects of pediatric intensive care sedation and analgesia: repetitive benzodiazepine and opioid exposure alters expression of glial and synaptic proteins in juvenile rats. Crit Care Explor. 2020;2(4):e0105. doi:10.1097/CCE.0000000000000105 [PubMed 32426747]
  105. Isbister GK, Calver LA, Downes MA, Page CB. Ketamine as rescue treatment for difficult-to-sedate severe acute behavioral disturbance in the emergency department. Ann Emerg Med. 2016;67(5):581-587.e1. doi:10.1016/j.annemergmed.2015.11.028 [PubMed 26899459]
  106. Jabre P, Combes X, Lapostolle F, et al; KETASED Collaborative Study Group. Etomidate versus ketamine for rapid sequence intubation in acutely ill patients: a multicentre randomised controlled trial. Lancet. 2009;374(9686):293-300. doi:10.1016/S0140-6736(09)60949-1 [PubMed 19573904]
  107. Jobeir A, Galal MO, Bulbul ZR, Solymar L, Darwish A, Schmaltz AA. Use of low-dose ketamine and/or midazolam for pediatric cardiac catheterization. Pediatr Cardiol. 2003;24(3):236-243. doi:10.1007/s00246-002-0339-5 [PubMed 12545318]
  108. Kalsi SS, Wood DM, Dargan PI. The epidemiology and patterns of acute and chronic toxicity associated with recreational ketamine use. Emerg Health Threats J. 2011;4:7107. doi:10.3402/ehtj.v4i0.7107 [PubMed 24149025]
  109. Kannan TR, Saxena A, Bhatnagar S, Barry A. Oral ketamine as an adjuvant to oral morphine for neuropathic pain in cancer patients. J Pain Symptom Manage. 2002;23(1):60-65. doi:10.1016/s0885-3924(01)00373-6 [PubMed 11779670]
  110. Ketalar (ketamine hydrochloride) [prescribing information]. Chestnut Ridge, NY: Par Pharmaceutical; June 2022.
  111. Ketalar (ketamine hydrochloride) [product monograph]. Montreal, Quebec, Canada: ERFA Canada; October 2020.
  112. Ketamine hydrochloride injection [prescribing information]. Lake Forest, IL: Hospira Inc; January 2013.
  113. Khorsand SM. Maintenance of general anesthesia: overview. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. Accessed September 21, 2020. http://www.uptodate.com.
  114. Kitch BB. Out-of-hospital ketamine: review of a growing trend in patient care. J Am Coll Emerg Physicians Open. 2020;1(3):183-189. doi:10.1002/emp2.12023 [PubMed 33000033]
  115. Koruk S, Mizrak A, Gul R, Kilic E, Yendi F, Oner U. Dexmedetomidine-ketamine and midazolam-ketamine combinations for sedation in pediatric patients undergoing extracorporeal shock wave lithotripsy: a randomized prospective study. J Anesth. 2010;24(6):858-863. doi:10.1007/s00540-010-1023-1. [PubMed 20924617]
  116. Kramer AH. Early ketamine to treat refractory status epilepticus. Neurocrit Care. 2012;16(2):299-305. doi:10.1007/s12028-011-9668-7 [PubMed 22237581]
  117. Lago P, Garetti E, Merazzi D, et al. Guidelines for procedural pain in the newborn. Acta Paediatr. 2009;98(6):932-939. doi:10.1111/j.1651-2227.2009.01291.x [PubMed 19484828]
  118. Lahti AC, Koffel B, LaPorte D, et al. Subanesthetic doses of ketamine stimulate psychosis in schizophrenia. Neuropsychopharmacology. 1995;13(1):9-19. doi:10.1016/0893-133X(94)00131-I [PubMed 8526975]
  119. Lai Y, Wu S, Ni L, Chen Z, Li X, Yang S, Gui Y, Guan Z, Cai Z, Ye J. Ketamine-associated urinary tract dysfunction: an underrecognized clinical entity. Urol Int. 2012;89(1):93-6. doi: 10.1159/000338098. Epub 2012 Jun 16. PMID: 22710265. [PubMed 22710265]
  120. Legriel S, Oddo M, Brophy GM. What's new in refractory status epilepticus? Intensive Care Med. 2017;43(4):543-546. doi:10.1007/s00134-016-4501-6 [PubMed 27544138]
  121. Li M, Martinelli AN, Oliver WD, Wilkerson RG. Evaluation of ketamine for excited delirium syndrome in the adult emergency department. J Emerg Med. 2019;S0736-S4679(19)30802-9. doi:10.1016/j.jemermed.2019.09.019 [PubMed 31735659]
  122. Lim DK. Ketamine associated psychedelic effects and dependence. Singapore Med J. 2003;44(1):31-34. [PubMed 12762561]
  123. Lin C, Durieux ME. Ketamine and kids: an update. Paediatr Anaesth. 2005;15(2):91-97. doi:10.1111/j.1460-9592.2005.01475.x [PubMed 15675923]
  124. Lin PC, Lane HY, Lin CH. Spontaneous remission of ketamine withdrawal-related depression. Clin Neuropharmacol. 2016;39(1):51-52. doi:10.1097/WNF.0000000000000121 [PubMed 26757313]
  125. Lin SM, Liu K, Tsai SK, Lee TY. Rectal ketamine versus intranasal ketamine as premedicant in children. Ma Zui Xue Za Zhi. 1990;28(2):177-183. [PubMed 2215104]
  126. Linder LM, Ross CA, Weant KA. Ketamine for the acute management of excited delirium and agitation in the prehospital setting. Pharmacotherapy. 2018;38(1):139-151. doi:10.1002/phar.2060 [PubMed 29136301]
  127. List WF, Crumrine RS, Cascorbi HF, Weiss MH. Increased cerebrospinal fluid pressure after ketamine. Anesthesiology. 1972;36(1):98-99. doi:10.1097/00000542-197201000-00023 [PubMed 5006995]
  128. Little B, Chang T, Chucot L, et al. Study of ketamine as an obstetric anesthetic agent. Am J Obstet Gynecol. 1972;15:113(2):247-260. doi:10.1016/0002-9378(72)90774-0 [PubMed 4554554]
  129. Louon A, Reddy VG. Nasal midazolam and ketamine for paediatric sedation during computerised tomography. Acta Anaesthesiol Scand. 1994;38(3):259-261. doi:10.1111/j.1399-6576.1994.tb03885.x [PubMed 8023666]
  130. Loveday BA, Sindt J. Ketamine protocol for palliative care in cancer patients with refractory pain. J Adv Pract Oncol. 2015;6(6):555-561. [PubMed 27648345]
  131. Lyon F, Dabbs T, O'Meara M. Ketamine sedation during the treatment of retinopathy of prematurity. Eye (Lond). 2008;22(5):684-686. doi:10.1038/sj.eye.6702717 [PubMed 17417623]
  132. Malhotra AK, Pinals DA, Adler CM, et al. Ketamine-induced exacerbation of psychotic symptoms and cognitive impairment in neuroleptic-free schizophrenics. Neuropsychopharmacology. 1997;17(3):141-150. doi:10.1016/S0893-133X(97)00036-5 [PubMed 9272481]
  133. Malinovsky JM, Servin F, Cozian A, Lepage JY, Pinaud M. Ketamine and norketamine plasma concentrations after i.v., nasal and rectal administration in children. Br J Anaesth. 1996;77(2):203-207. doi:10.1093/bja/77.2.203 [PubMed 8881626]
  134. Marchetti F, Coutaux A, Bellanger A, Magneux C, Bourgeois P, Mion G. Efficacy and safety of oral ketamine for the relief of intractable chronic pain: A retrospective 5-year study of 51 patients. Eur J Pain. 2015;19(7):984-993. doi:10.1002/ejp.624 [PubMed 25381898]
  135. Martin E, Vickers B, Landau R, Reece-Stremtan S. ABM clinical protocol #28, peripartum analgesia and anesthesia for the breastfeeding mother. Breastfeed Med. 2018;13(3):164-171. doi:10.1089/bfm.2018.29087.ejm [PubMed 29595994]
  136. McGirr A, Berlim MT, Bond DJ, Fleck MP, Yatham LN, Lam RW. A systematic review and meta-analysis of randomized, double-blind, placebo-controlled trials of ketamine in the rapid treatment of major depressive episodes. Psychol Med. 2015;45(4):693-704. doi:10.1017/S0033291714001603 [PubMed 25010396]
  137. McGlone RG, Howes MC, Joshi M. The Lancaster Experience of 2.0 to 2.5 mg/kg Intramuscular Ketamine for Paediatric Sedation: 501 Cases and Analysis. Emerg Med J. 2004;21(3):290-295. doi:10.1136/emj.2002.003772 [PubMed 15107365]
  138. McPherson C. Sedation and analgesia in mechanically ventilated preterm neonates: continue standard of care or experiment? J Pediatr Pharmacol Ther. 2012;17(4):351-364. doi:10.5863/1551-6776-17.4.351 [PubMed 23413121]
  139. Melendez E, Bachur R. Serious adverse events during procedural sedation with ketamine. Pediatr Emerg Care. 2009;25(5):325-328. doi:10.1097/PEC.0b013e3181a341e0 [PubMed 19404223]
  140. Mellon RD, Simone AF, Rappaport BA. Use of anesthetic agents in neonates and young children. Anesth Analg. 2007;104(3):509-520. doi:10.1213/01.ane.0000255729.96438.b0 [PubMed 17312200]
  141. Mercadante S, Arcuri E, Tirelli W, Casuccio A. Analgesic effect of intravenous ketamine in cancer patients on morphine therapy: a randomized, controlled, double-blind, crossover, double-dose study. J Pain Symptom Manage. 2000;20(4):246-252. doi:10.1016/s0885-3924(00)00194-9 [PubMed 11027905]
  142. Mercadante S, Caruselli A, Casuccio A. The use of ketamine in a palliative-supportive care unit: a retrospective analysis. Ann Palliat Med. 2018;7(2):205-210. doi:10.21037/apm.2018.01.01 [PubMed 29764182]
  143. Miller M, Kruit N, Heldreich C, et al. Hemodynamic response after rapid sequence induction with ketamine in out-of-hospital patients at risk of shock as defined by the shock index. Ann Emerg Med. 2016;68(2):181-188.e2. doi:10.1016/j.annemergmed.2016.03.041 [PubMed 27130803]
  144. Miller RD. Miller’s Anesthesia. 7th ed. Churchill Livingstone; 2010.
  145. Moore G, Pfaff JA. Assessment and emergency management of the acutely agitated or violent adult. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. Accessed July 29, 2020. http://www.uptodate.com.
  146. Moore NN, Bostwick JM. Ketamine dependence in anesthesia providers. Psychosomatics. 1999;40(4):356-359. doi:10.1016/S0033-3182(99)71231-4 [PubMed 10402883]
  147. Murrough JW, Perez AM, Pillemer S, et al. Rapid and longer-term antidepressant effects of repeated ketamine infusions in treatment-resistant major depression. Biol Psychiatry. 2013;74(4):250-256. doi:10.1016/j.biopsych.2012.06.022 [PubMed 22840761]
  148. Nagdeve NG, Yaddanapudi S, Pandav SS. The effect of different doses of ketamine on intraocular pressure in anesthetized children. J Pediatr Ophthalmol Strabismus. 2006;43(4):219-223. doi:10.3928/01913913-20060701-03 [PubMed 16915900]
  149. Nazarian DJ, Broder JS, Thiessen MEW, Wilson MP, Zun LS, Brown MD; American College of Emergency Physicians Clinical Policies Subcommittee (Writing Committee) on the Adult Psychiatric Patient. Clinical policy: critical issues in the diagnosis and management of the adult psychiatric patient in the emergency department. Ann Emerg Med. 2017;69(4):480-498. doi:10.1016/j.annemergmed.2017.01.036. [PubMed 28335913]
  150. Neuman G, Koren G. Safety of procedural sedation in pregnancy. J Obstet Gynaecol Can. 2013;35(2):168-173. doi:10.1016/S1701-2163(15)31023-9. [PubMed 23470068]
  151. Newport DJ, Carpenter LL, McDonald WM, Potash JB, Tohen M, Nemeroff CB; APA Council of Research Task Force on Novel Biomarkers and Treatments. Ketamine and other NMDA antagonists: early clinical trials and possible mechanisms in depression. Am J Psychiatry. 2015;172(10):950-966. doi:10.1176/appi.ajp.2015.15040465 [PubMed 26423481]
  152. Nielsen BN, Friis SM, Rømsing J, et al. Intranasal sufentanil/ketamine analgesia in children. Paediatr Anaesth. 2014;24(2):170-180. doi:10.1111/pan.12268 [PubMed 24118506]
  153. Niesters M, Martini C, Dahan A. Ketamine for chronic pain: Risks and benefits. Br J Clin Pharmacology. 2013;77(2):357-367. doi:10.1111/bcp.12094
  154. Norambuena C, Yañez J, Flores V, Puentes P, Carrasco P, Villena R. Oral ketamine and midazolam for pediatric burn patients: a prospective, randomized, double-blind study. J Pediatr Surg. 2013;48(3):629-634. doi:10.1016/j.jpedsurg.2012.08.018. [PubMed 23480923]
  155. O'Brien ME, Fuh L, Raja AS, White BA, Yun BJ, Hayes BD. Reduced-dose intramuscular ketamine for severe agitation in an academic emergency department. Clin Toxicol (Phila). 2020;58(4):294-298. doi:10.1080/15563650.2019.1643468 [PubMed 31335216]
  156. Oddo M, Bracard S, Cariou A, et al. Update in neurocritical care: a summary of the 2018 Paris international conference of the French Society of Intensive Care. Ann Intensive Care. 2019;9(1):47. doi:10.1186/s13613-019-0523-x [PubMed 30993550]
  157. Okon T. Ketamine: an introduction for the pain and palliative medicine physician. Pain Physician. 2007;10(3):493-500. [PubMed 17525784]
  158. Oliveira J E Silva L, Lee JY, Bellolio F, Homme JL, Anderson JL. Intranasal ketamine for acute pain management in children: A systematic review and meta-analysis. Am J Emerg Med. 2020;38(9):1860-1866. doi:10.1016/j.ajem.2020.05.094 [PubMed 32739857]
  159. Olutoye OA, Baker BW, Belfort MA, Olutoye OO. Food and Drug Administration warning on anesthesia and brain development: implications for obstetric and fetal surgery. Am J Obstet Gynecol. 2018;218(1):98-102. doi:10.1016/j.ajog.2017.08.107 [PubMed 28888583]
  160. Orhurhu V, Orhurhu MS, Bhatia A, Cohen SP. Ketamine infusions for chronic pain: a systematic review and meta-analysis of randomized controlled trials. Anesth Analg. 2019;129(1):241-254. doi:10.1213/ANE.0000000000004185 [PubMed 31082965]
  161. Ozdemir D, Kayserili E, Arslanoglu S, Gulez P, Vergin C. Ketamine and midazolam for invasive procedures in children with malignancy: a comparison of routes of intravenous, oral, and rectal administration. J Trop Pediatr. 2004;50(4):224-228. doi:10.1093/tropej/50.4.224 [PubMed 15357562]
  162. Page RL 2nd, O'Bryant CL, Cheng D, et al; American Heart Association Clinical Pharmacology and Heart Failure and Transplantation Committees of the Council on Clinical Cardiology; Council on Cardiovascular Surgery and Anesthesia; Council on Cardiovascular and Stroke Nursing; and Council on Quality of Care and Outcomes Research. Drugs That May Cause or Exacerbate Heart Failure: A Scientific Statement From the American Heart Association [published correction appears in Circulation. 2016;134(12):e261]. Circulation. 2016;134(6):e32-e69. doi:10.1161/CIR.0000000000000426 [PubMed 27400984]
  163. Pai A, Heining M. Ketamine. Continuing education in anaesthesia, critical care and pain. Br J Anaesthesia. 2007;7(2):59-63. Accessed December 2021. https://academic.oup.com/bjaed/article/7/2/59/384361.
  164. Pal HR, Berry N, Kumar R, Ray R. Ketamine dependence. Anaesth Intensive Care. 2002;30(3):382-384. doi:10.1177/0310057X0203000323 [PubMed 12075653]
  165. Pal R, Balt S, Erowid E, et al. Ketamine is associated with lower urinary tract signs and symptoms. Drug Alcohol Depend. 2013;132(1-2):189-194. doi:10.1016/j.drugalcdep.2013.02.005 [PubMed 23474358]
  166. Patel P, Sun L. Update on neonatal anesthetic neurotoxicity: insight into molecular mechanisms and relevance to humans. Anesthesiology. 2009;110(4):703-708. doi:10.1097/ALN.0b013e31819c42a4 [PubMed 19276968]
  167. Pees C, Haas NA, Ewert P, Berger F, Lange PE. Comparison of analgesic/sedative effect of racemic ketamine and S(+)-ketamine during cardiac catheterization in newborns and children. Pediatr Cardiol. 2003;24(5):424-429. doi:10.1007/s00246-002-0356-4 [PubMed 14627307]
  168. Peng TR, Lee MC, Wu TW, Lan CC. Suspected ketamine-associated lower urinary tract symptoms. Urol J. 2014;11(2):1508-1510. [PubMed 24807773]
  169. Phillips JL, Norris S, Talbot J, et al. Single and repeated ketamine infusions for reduction of suicidal ideation in treatment-resistant depression. Neuropsychopharmacology. 2020;45(4):606-612. doi:10.1038/s41386-019-0570-x [PubMed 31759333]
  170. Phillips JL, Norris S, Talbot J, et al. Single, repeated, and maintenance ketamine infusions for treatment-resistant depression: a randomized controlled trial. Am J Psychiatry. 2019;176(5):401-409. doi:10.1176/appi.ajp.2018.18070834 [PubMed 30922101]
  171. Poonai N, Canton K, Ali S, et al. Intranasal ketamine for procedural sedation and analgesia in children: A systematic review. PLoS One. 2017;12(3):e0173253. doi:10.1371/journal.pone.0173253 [PubMed 28319161]
  172. Powers AR 3rd, Gancsos MG, Finn ES, Morgan PT, Corlett PR. Ketamine-induced hallucinations. Psychopathology. 2015;48(6):376-385. doi:10.1159/000438675 [PubMed 26361209]
  173. Quibell R, Fallon M, Mihalyo M, Twycross R, Wilcock A. Ketamine. J Pain Symptom Manage. 2015;50(2):268-278. doi:10.1016/j.jpainsymman.2015.06.002 [PubMed 26096492]
  174. Quibell R, Prommer EE, Mihalyo M, Twycross R, Wilcock A. Ketamine*. J Pain Symptom Manage. 2011;41(3):640-649. doi:10.1016/j.jpainsymman.2011.01.001 [PubMed 21419322]
  175. Rai S, Drislane FW. Treatment of refractory and super-refractory status epilepticus. Neurotherapeutics. 2018;15(3):697-712. doi:10.1007/s13311-018-0640-5 [PubMed 29922905]
  176. Rasmussen KG, Lineberry TW, Galardy CW, et al. Serial infusions of low-dose ketamine for major depression. J Psychopharmacol. 2013;27(5):444-450. doi:10.1177/0269881113478283. [PubMed 23428794]
  177. Rech MA, Barbas B, Chaney W, Greenhalgh E, Turck C. When to pick the nose: out-of-hospital and emergency department intranasal administration of medications. Ann Emerg Med. 2017;70(2):203-211. doi:10.1016/j.annemergmed.2017.02.015. [PubMed 28366351]
  178. Reece-Stremtan S, Campos M, Kokajko L; Academy of Breastfeeding Medicine. ABM clinical protocol #15: analgesia and anesthesia for the breastfeeding mother, revised 2017. Breastfeed Med. 2017;12(9):500-506. doi:10.1089/bfm.2017.29054.srt [PubMed 29624435]
  179. Reynolds SL, Bryant KK, Studnek JR, et al. Randomized controlled feasibility trial of intranasal ketamine compared to intranasal fentanyl for analgesia in children with suspected extremity fractures. Acad Emerg Med. 2017;24(12):1430-1440. doi:10.1111/acem.13313 [PubMed 28926159]
  180. Riddell J, Tran A, Bengiamin R, Hendey GW, Armenian P. Ketamine as a first-line treatment for severely agitated emergency department patients. Am J Emerg Med. 2017;35(7):1000-1004. doi:10.1016/j.ajem.2017.02.026. [PubMed 28237385]
  181. Roberts JR, Custalow CB, Thomsenet TW, et al, eds. Roberts and Hedges' clinical procedures in emergency medicine and acute care, 7th ed. Elsevier. 2019.
  182. Rock MJ, Reyes de la Rocha S, L'Hommedieu CS, Truemper E. Use of ketamine in asthmatic children to treat respiratory failure refractory to conventional therapy. Crit Care Med. 1986;14(5):514-516. [PubMed 3698618]
  183. Roelofse JA, Shipton EA, de la Harpe CJ, Blignaut RJ. Intranasal sufentanil/midazolam versus ketamine/midazolam for analgesia/sedation in the pediatric population prior to undergoing multiple dental extractions under general anesthesia: a prospective, double-blind, randomized comparison. Anesth Prog. 2004;51(4):114-121. [PubMed 15675259]
  184. Rosati A, De Masi S, Guerrini R. Ketamine for refractory status epilepticus: a systematic review. CNS Drugs. 2018;32(11):997-1009. doi:10.1007/s40263-018-0569-6 [PubMed 30232735]
  185. Sanacora G, Frye MA, McDonald W, et al. A consensus statement on the use of ketamine in the treatment of mood disorders. JAMA Psychiatry. 2017;74(4):399-405. doi:10.1001/jamapsychiatry.2017.0080 [PubMed 28249076]
  186. Schumann R. Anesthesia for the obese patient. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. Accessed July 29, 2020. http://www.uptodate.com.
  187. Schwenk ES, Viscusi ER, Buvanendran A, et al. Consensus guidelines on the use of intravenous ketamine infusions for acute pain management from the American Society of Regional Anesthesia and Pain Medicine, the American Academy of Pain Medicine, and the American Society of Anesthesiologists. Reg Anesth Pain Med. 2018;43(5):456-466. doi:10.1097/AAP.0000000000000806 [PubMed 29870457]
  188. Shah A, Mosdossy G, McLeod S, Lehnhardt K, Peddle M, Rieder M. A blinded, randomized controlled trial to evaluate ketamine/propofol versus ketamine alone for procedural sedation in children. Ann Emerg Med. 2011;57(5):425-433.e2. doi:10.1016/j.annemergmed.2010.08.032. [PubMed 20947210]
  189. Shimonovich S, Gigi R, Shapira A, et al. Intranasal ketamine for acute traumatic pain in the emergency department: a prospective, randomized clinical trial of efficacy and safety. BMC Emerg Med. 2016;16(1):43. doi:10.1186/s12873-016-0107-0 [PubMed 27829367]
  190. Shiroma PR, Johns B, Kuskowski M, et al. Augmentation of response and remission to serial intravenous subanesthetic ketamine in treatment resistant depression. J Affect Disord. 2014;155:123-129. doi: 10.1016/j.jad.2013.10.036. [PubMed 24268616]
  191. Shrestha R, Pant S, Shrestha A, Batajoo KH, Thapa R, Vaidya S. Intranasal ketamine for the treatment of patients with acute pain in the emergency department. World J Emerg Med. 2016;7(1):19-24. doi:10.5847/wjem.j.1920-8642.2016.01.003 [PubMed 27006733]
  192. Singh JB, Fedgchin M, Daly EJ, et al. A double-blind, randomized, placebo-controlled, dose-frequency study of intravenous ketamine in patients with treatment-resistant depression. Am J Psychiatry. 2016;173(8):816-826. doi:10.1176/appi.ajp.2016.16010037 [PubMed 27056608]
  193. Singh V, Gillespie TW, Harvey RD. Intranasal ketamine and its potential role in cancer-related pain. Pharmacotherapy. 2018;38(3):390-401. doi:10.1002/phar.2090 [PubMed 29396996]
  194. Soto E, Stewart DR, Mannes AJ, et al. Oral ketamine in the palliative care setting: a review of the literature and case report of a patient with neurofibromatosis type 1 and glomus tumor-associated complex regional pain syndrome. Am J Hosp Palliat Care. 2012;29(4): 308–317. doi:10.1177/1049909111416345 [PubMed 21803784]
  195. Society for Pediatric Sedation (SPS). FDA advisory on anesthesia and sedation medication in children. Society for Pediatric Sedation website. Accessed April 2021. https://www.pedsedation.org/resources/quality-safety/fda-advisory/#:~:text=In%20December%202016%2C%20the%20FDA,(http%3A%2F%2Fwww.fda.
  196. Stollings JL, Diedrich DA, Oyen LJ, Brown DR. Rapid-sequence intubation: a review of the process and considerations when choosing medications. Ann Pharmacother. 2014;48(1):62-76. doi:10.1177/1060028013510488 [PubMed 24259635]
  197. Sungur Ulke Z, Kartal U, Orhan Sungur M, Camci E, Tugrul M. Comparison of sevoflurane and ketamine for anesthetic induction in children with congenital heart disease. Paediatr Anaesth. 2008;18(8):715-21. doi:10.1111/j.1460-9592.2008.02637.x. [PubMed 18544145]
  198. Surendar MN, Pandey RK, Saksena AK, Kumar R, Chandra G. A comparative evaluation of intranasal dexmedetomidine, midazolam and ketamine for their sedative and analgesic properties: a triple blind randomized study. J Clin Pediatr Dent. 2014;38(3):255-261. doi:10.17796/jcpd.38.3.l828585807482966 [PubMed 25095322]
  199. Swainson J, McGirr A, Blier P, et al. The Canadian Network for Mood and Anxiety Treatments (CANMAT) task force recommendations for the use of racemic ketamine in adults with major depressive disorder: recommandations Du Groupe De Travail Du Réseau Canadien Pour Les Traitements De L'humeur Et De L'anxiété (Canmat) Concernant L'utilisation De La Kétamine Racémique Chez Les Adultes Souffrant De Trouble Dépressif Majeur. Can J Psychiatry. 2021;66(2):113-125. doi:10.1177/0706743720970860 [PubMed 33174760]
  200. Szarmach J, Cubała WJ, Włodarczyk A, Wiglusz MS. Short-term ketamine administration in treatment-resistant depression: focus on cardiovascular safety. Psychiatr Danub. 2019;31(suppl 3):585-590. [PubMed 31488795]
  201. Talahma M, Sabharwal V, Bukovskaya Y, Khan F. Ketamine infusion used to successfully control refractory status epilepticus in a pregnant patient. Case Rep Neurol Med. 2018;2018:3041279. doi:10.1155/2018/3041279 [PubMed 30498609]
  202. Tanaka M, Sato M, Saito A, Nishikawa T. Reevaluation of rectal ketamine premedication in children: comparison with rectal midazolam. Anesthesiology. 2000;93(5):1217-1224. doi:10.1097/00000542-200011000-00014 [PubMed 11046209]
  203. Tawfic QA, Faris AS, Kausalya R. The role of a low-dose ketamine-midazolam regimen in the management of severe painful crisis in patients with sickle cell disease. J Pain Symptom Manage. 2014;47(2):334-340. doi:10.1016/j.jpainsymman.2013.03.012 [PubMed 23856095]
  204. Thase M, Connolly KR. Ketamine and esketamine for treating unipolar depression in adults: administration, efficacy, and adverse effects. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. Accessed March 5, 2020. http://www.uptodate.com.
  205. Tobias JD, Martin LD, Wetzel RC. Ketamine by continuous infusion for sedation in the pediatric intensive care unit. Crit Care Med. 1990;18(8):819-821. doi:10.1097/00003246-199008000-00004 [PubMed 2379394]
  206. Tobias JD, Phipps S, Smith B, Mulhern RK. Oral ketamine premedication to alleviate the distress of invasive procedures in pediatric oncology patients. Pediatrics. 1992;90(4):537-541. [PubMed 1408506]
  207. Tobias JD, Rasmussen GE. Pain management and sedation in the pediatric intensive care unit. Pediatr Clin North Am. 1994;41(6):1269-1292. doi:10.1016/s0031-3955(16)38873-3 [PubMed 7984386]
  208. Tomatir E, Atalay H, Gurses E, Erbay H, Bozkurt P. Effects of low dose ketamine before induction on propofol anesthesia for pediatric magnetic resonance imaging. Paediatr Anaesth. 2004;14(10):845-850. doi:10.1111/j.1460-9592.2004.01303.x [PubMed 15385013]
  209. Turhanoğlu S, Kararmaz A, Ozyilmaz MA, Kaya S, Tok D. Effects of different doses of oral ketamine for premedication of children. Eur J Anaesthesiol. 2003;20(1):56-60. doi:10.1017/s0265021503000103 [PubMed 12553389]
  210. Upchurch CP, Grijalva CG, Russ S, et al. Comparison of etomidate and ketamine for induction during rapid sequence intubation of adult trauma patients. Ann Emerg Med. 2017;69(1):24-33.e2. doi:10.1016/j.annemergmed.2016.08.009 [PubMed 27993308]
  211. US Department of Veterans Affairs/Department of Defense (VA/DoD). VA/DoD clinical practice guideline for the assessment and management of patients as risk for suicide. Updated May 2019. Accessed March 24, 2020. https://www.healthquality.va.gov/guidelines/MH/srb/VADoDSuicideRiskFullCPGFinal5088919.pdf.
  212. Vadivelu N, Schermer E, Kodumudi V, Belani K, Urman RD, Kaye AD. Role of ketamine for analgesia in adults and children. J Anaesthesiol Clin Pharmacol. 2016;32(3):298-306. doi:10.4103/0970-9185.168149 [PubMed 27625475]
  213. van der Bijl P, Roelofse JA, Stander IA. Rectal ketamine and midazolam for premedication in pediatric dentistry. J Oral Maxillofac Surg. 1991;49(10):1050-1054. doi:10.1016/0278-2391(91)90136-a [PubMed 1890517]
  214. Wagner BK, O'Hara DA. Pharmacokinetics and pharmacodynamics of sedatives and analgesics in the treatment of agitated critically ill patients. Clin Pharmacokinet. 1997;33(6):426-453. doi:10.2165/00003088-199733060-00003 [PubMed 9435992]
  215. Wang J, Echevarria GC, Doan L, et al. Effects of a single subanaesthetic dose of ketamine on pain and mood after laparoscopic bariatric surgery: a randomised double-blind placebo controlled study. Eur J Anaesthesiol. 2019;36(1):16-24. doi:10.1097/EJA.0000000000000860 [PubMed 30095550]
  216. Wang X, Zhou ZJ, Zhang XF, Zheng S. A comparison of two different doses of rectal ketamine added to 0.5 mg x kg(-1) midazolam and 0.02 mg x kg(-1) atropine in infants and young children. Anaesth Intensive Care. 2010;38(5):900-904. doi:10.1177/0310057X1003800515 [PubMed 20865876]
  217. Wei YB, Yang JR, Yin Z, Guo Q, Liang BL, Zhou KQ. Genitourinary toxicity of ketamine. Hong Kong Med J. 2013;19(4):341-348. doi:10.12809/hkmj134013 [PubMed 23832948]
  218. Weksler N, Ovadia L, Muati G, Stav A. Nasal ketamine for paediatric premedication. Can J Anaesth. 1993;40(2):119-121. doi:10.1007/BF03011307 [PubMed 8443849]
  219. White PF, Way WL, Trevor AJ. Ketamine − its pharmacology and therapeutic uses. Anesthesiology. 1982;56(2):119-136. doi:10.1097/00000542-198202000-00007 [PubMed 6892475]
  220. 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(1):23-30. doi:10.1016/j.annemergmed.2006.08.002 [PubMed 17059854]
  221. Witt K, Potts J, Hubers A, et al. Ketamine for suicidal ideation in adults with psychiatric disorders: a systematic review and meta-analysis of treatment trials. Aust N Z J Psychiatry. 2020;54(1):29-45. doi:10.1177/0004867419883341 [PubMed 31729893]
  222. Wu L, Lalwani K, Hook KA, Almario BM, Fu R, Edmunds B. Respiratory complications associated with ketamine anesthesia for ophthalmic procedures following intraocular pressure measurement in children. J Anaesthesiol Clin Pharmacol. 2014;30(2):253-257. doi:10.4103/0970-9185.130047 [PubMed 24803768]
  223. Xiong J, Lipsitz O, Chen-Li D, et al. The acute antisuicidal effects of single-dose intravenous ketamine and intranasal esketamine in individuals with major depression and bipolar disorders: a systematic review and meta-analysis. J Psychiatr Res. 2021;134:57-68. doi:10.1016/j.jpsychires.2020.12.038 [PubMed 33360864]
  224. Yan J, Li YR, Zhang Y, Lu Y, Jiang H. Repeated exposure to anesthetic ketamine can negatively impact neurodevelopment in infants: a prospective preliminary clinical study. J Child Neurol. 2014;29(10):1333-1338. doi:10.1177/0883073813517508 [PubMed 24659739]
  225. Yeaman F, Meek R, Egerton-Warburton D, Rosengarten P, Graudins A. Sub-dissociative-dose intranasal ketamine for moderate to severe pain in adult emergency department patients. Emerg Med Australas. 2014;26(3):237-242. doi:10.1111/1742-6723.12173 [PubMed 24712757]
  226. Yenigun A, Yilmaz S, Dogan R, Goktas SS, Calim M, Ozturan O. Demonstration of analgesic effect of intranasal ketamine and intranasal fentanyl for postoperative pain after pediatric tonsillectomy. Int J Pediatr Otorhinolaryngol. 2018;104:182-185. doi:10.1016/j.ijporl.2017.11.018 [PubMed 29287863]
  227. Youssef-Ahmed MZ, Silver P, Nimkoff L, Sagy M. Continuous infusion of ketamine in mechanically ventilated children with refractory bronchospasm. Intensive Care Med. 1996;22(9):972-976. doi:10.1007/BF02044126 [PubMed 8905436]
  228. Zanette G, Micaglio M, Zanette L, Manani G, Facco E. Comparison between ketamine and fentanyl-droperidol for rectal premedication in children: a randomized placebo controlled trial. J Anesth. 2010;24(2):197-203. doi:10.1007/s00540-010-0884-7. [PubMed 20157833]
  229. Zeiler FA, Teitelbaum J, West M, Gillman LM. The ketamine effect on ICP in traumatic brain injury. Neurocrit Care. 2014;21(1):163-173. doi:10.1007/s12028-013-9950-y. [PubMed 24515638]
Topic 13398 Version 369.0