Your activity: 18 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: sshnevis@outlook.com

Tramadol: Drug information

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

For abbreviations, symbols, and age group definitions used in Lexicomp (show table)
ALERT: US Boxed Warning
Risk of medication errors:

Ensure accuracy when prescribing, dispensing, and administering tramadol oral solution. Dosing errors due to confusion between mg and mL can result in accidental overdose and death.

Addiction, abuse, and misuse:

Tramadol exposes patients and other users to the risks of opioid addiction, abuse, and misuse, which can lead to overdose and death. Assess each patient's risk prior to prescribing tramadol, and monitor all patients regularly for the development of these behaviors and conditions.

Opioid analgesic Risk Evaluation and Mitigation Strategy (REMS):

To ensure that the benefits of opioid analgesics outweigh the risks of addiction, abuse, and misuse, the Food and Drug Administration (FDA) has required a REMS for these products. Under the requirements of the REMS, drug companies with approved opioid analgesic products must make REMS-compliant education programs available to healthcare providers. Healthcare providers are strongly encouraged to complete a REMS-compliant education program; counsel patients and/or their caregivers, with every prescription, on safe use, serious risks, storage, and disposal of these products; emphasize to patients and their caregivers the importance of reading the Medication Guide every time it is provided by their pharmacist; and consider other tools to improve patient, household, and community safety.

Life-threatening respiratory depression:

Serious, life-threatening, or fatal respiratory depression may occur with use of tramadol. Monitor for respiratory depression, especially during initiation of tramadol or following a dose increase. Instruct patients to swallow tramadol capsules and extended-release tablets intact, and not to split, break, chew, crush, or dissolve the contents of the capsules or extended-release tablets to avoid exposure to a potentially fatal dose of tramadol.

Accidental ingestion:

Accidental ingestion of tramadol, especially by children, can be fatal.

Ultra-rapid metabolism of tramadol and other risk factors for life-threatening respiratory depression in children:

Life-threatening respiratory depression and death have occurred in children who received tramadol. Some of the reported followed tonsillectomy and/or adenoidectomy; in at least 1 case, the child had evidence of being an ultra-rapid metabolizer of tramadol due to a CYP450 2D6 polymorphism. Tramadol is contraindicated in children <12 years of age and in children <18 years of age following tonsillectomy and/or adenoidectomy. Avoid the use of tramadol in adolescents 12 to 18 years of age who have other risk factors that may increase their sensitivity to the respiratory depressant effects of tramadol.

Neonatal opioid withdrawal syndrome:

Prolonged use of tramadol during pregnancy can result in neonatal opioid withdrawal syndrome, which may be life-threatening if not recognized and treated, and requires management according to protocols developed by neonatology experts. If opioid use is required for a prolonged period in a pregnant woman, advise the patient of the risk of neonatal opioid withdrawal syndrome and ensure that appropriate treatment will be available.

Interactions with drugs affecting cytochrome P450 isoenzymes:

The effects of concomitant use or discontinuation of cytochrome P450 3A4 inducers, 3A4 inhibitors, or 2D6 inhibitors with tramadol are complex. Use of cytochrome P450 3A4 inducers, 3A4 inhibitors, or 2D6 inhibitors with tramadol requires careful consideration of the effects on the parent drug, tramadol, and the active metabolite, M1.

Risks from concomitant use with benzodiazepines or other CNS depressants:

Concomitant use of opioids with benzodiazepines or other CNS depressants, including alcohol, may result in profound sedation, respiratory depression, coma, and death. Reserve concomitant prescribing of tramadol and benzodiazepines or other CNS depressants for use in patients for whom alternative treatment options are inadequate. Limit treatment to the minimum effective dosages and durations. Follow patients for signs and symptoms of respiratory depression and sedation.

Brand Names: US
  • ConZip;
  • Qdolo;
  • Ultram [DSC]
Brand Names: Canada
  • APO-Tramadol;
  • AURO-Tramadol;
  • Durela;
  • JAMP Tramadol;
  • MAR-Tramadol;
  • Ralivia;
  • SANDOZ Tramadol [DSC];
  • TARO-Tramadol ER;
  • Tridural;
  • Ultram [DSC];
  • Zytram XL
Pharmacologic Category
  • Analgesic, Opioid
Dosing: Adult
Pain management, moderate to severe

Pain management, moderate to severe:

Note: In general, opioids may be considered a potential component of a comprehensive, multimodal, patient-specific treatment plan for pain. Nonopioid analgesia should be maximized, if appropriate, prior to initiation of opioid analgesia; combination therapy with analgesics with differing mechanisms of action may improve efficacy and reduce the doses and/or frequency required for each agent (Ref). Tramadol doses should be titrated to appropriate analgesic effect; use the lowest effective dose for the shortest period of time. Tramadol is used for a variety of moderate to moderately severe painful conditions and may be of particular benefit for patients with mixed nociceptive and neuropathic pain due to its dual mechanism of action (Ref).

Acute pain (eg, postoperative):

Note: In patients who are experiencing acute pain severe enough to require opioids (in addition to appropriate nonopioid analgesia), limit the quantity prescribed to the expected duration of acute pain; a quantity sufficient for ≤3 days is often adequate, whereas >7 days is rarely needed (Ref). Long-acting preparations are not recommended for treatment of acute pain in opioid-naive patients (Ref). Some experts avoid the use of tramadol in patients with moderate to severe acute pain due to the wide interpatient variability in metabolism and related incidences of adverse events and unreliable analgesia (Ref).

Immediate release: Oral: Initial: 50 mg every 4 to 6 hours as needed (Ref); some experts suggest that 25 to 50 mg 3 times per day may be sufficient for patients with moderate acute pain (Ref). The dose may be increased as needed and tolerated to 50 to 100 mg every 4 to 6 hours (maximum: 400 mg/day) (Ref).

Chronic pain (alternative agent):

Note: Opioids, including tramadol, are not the preferred therapy for chronic noncancer pain due to insufficient evidence of benefit and risk of serious harm; nonpharmacologic treatment and nonopioid analgesics are preferred with the exception of pain from sickle cell disease and in end-of-life care (Ref). Opioids, including tramadol, should only be considered in patients who experience clinically meaningful improvement in pain and function that outweighs patient safety risks (Ref). The utility of tramadol in patients with chronic pain due to cancer is questionable, especially considering its dual mechanism of action and dose ceiling (Ref).

Opioid-naive patients not currently on tramadol immediate release:

Immediate release: Oral: The ideal dosing regimen has not been established; consider restricting the initial dose to <300 mg tramadol per day (ie, <50 mg morphine equivalents daily) (Ref). An example initial dose is 25 to 50 mg every 6 hours as needed (Rosenquist 2022). The dose may be increased as needed and tolerated to 50 to 100 mg every 4 to 6 hours (maximum: 400 mg/day) (Ref).

Extended release:

Note: Although manufacturer's labeling contains the following directions for initiating ER tramadol products in opioid-naive patients with chronic pain, it is recommended that when starting opioid therapy, treatment be initiated with an IR preparation to more accurately determine the daily opioid requirement and decrease the risk of overdose (Ref). The CDC recommends that ER opioids be reserved for patients who have received IR opioids daily for ≥1 week yet continue to experience severe, continuous pain (Ref).

Initial: Oral: 100 mg once daily; titrate by 100 mg/day increments every 5 days as needed (maximum: 300 mg/day)

Tridural [Canadian product]: Initial: Oral: 100 mg once daily; titrate by 100 mg/day increments every 2 days as needed (maximum: 300 mg/day)

Zytram XL [Canadian product]: Oral: 150 mg once daily; if pain relief is not achieved, may titrate by increasing dosage incrementally with sufficient time to evaluate effect of increased dosage, generally not more often than every 7 days (maximum: 400 mg/day).

Patients currently on tramadol IR tablets for ≥1 week: Calculate 24-hour tramadol IR tablet total dose and initiate total ER daily dose (round dose to the next lowest 100 mg increment); titrate as needed and tolerated to desired effect (maximum: 300 mg/day). In patients who experience breakthrough pain, clinicians may consider the addition of an IR rescue analgesic (eg, NSAID or short-acting weak opioid). Note: Oral solution relative bioavailability compared to ER products has not been established; conversion to ER products should be accompanied by close monitoring of excessive sedation and respiratory depression.

Discontinuation or tapering of therapy: When discontinuing or tapering chronic opioid therapy, the dose should be gradually tapered down. An optimal universal tapering schedule for all patients has not been established (Ref). Proposed schedules range from slow (eg, 10% reductions per week or 10% reduction per month depending on duration of chronic therapy) to rapid (eg, 25% to 50% reduction every few days) (Ref). Individualize based on discussions with patient to minimize opioid withdrawal while considering patient-specific goals and concerns as well as the pharmacokinetics of the opioid being tapered. Slower tapers may be appropriate in patients who have been receiving opioids for a long duration (eg, years), particularly in the final stage of tapering, whereas more rapid tapers may be appropriate in patients experiencing severe adverse events (Ref). Monitor carefully for signs/symptoms of withdrawal. If the patient displays withdrawal symptoms, consider slowing the taper schedule; alterations may include increasing the interval between dose reductions, decreasing amount of daily dose reduction, pausing the taper and restarting when the patient is ready, and/or coadministration of an alpha-2 agonist (eg, clonidine) to blunt withdrawal symptoms (Ref). Continue to offer nonopioid analgesics as needed for pain management during the taper; consider nonopioid adjunctive treatments for withdrawal symptoms (eg, GI complaints, muscle spasm) as needed (Ref).

Premature ejaculation

Premature ejaculation (alternative agent) (off-label use):

Note: Tramadol may be considered in patients who have failed other therapies (eg, SSRIs, topical anesthetics). Consideration should be given to the risk of addiction and adverse effects associated with opioids (Ref); to promote safe use, regular follow-up to monitor for response, toxicity, and misuse is recommended.

Immediate release: Oral: The ideal dosing regimen has not been established; dosage range studied: 25 to 50 mg administered on demand 1 to 3 hours prior to intercourse (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

The renal dosing recommendations are based upon the best available evidence and clinical expertise. Senior Editorial Team: Bruce Mueller, PharmD, FCCP, FASN, FNKF; Jason Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC; Michael Heung, MD, MS.

Altered kidney function (Ref):

CrCl ≥30 mL/minute: Immediate release, extended release: No dosage adjustment necessary.

CrCl <30 mL/minute: Immediate release: Increase dosing interval to every 12 hours; maximum: 200 mg/day. ER formulation should be avoided.

Hemodialysis, intermittent (thrice weekly): Dialyzable (7%):

Immediate release: Lower initial doses and an extended dosing interval (eg, 25 mg twice daily) are recommended (Ref); titrate to response. The manufacturer’s labeling recommends a maximum daily dose of 200 mg/day; however, since a uremic state may lower seizure threshold, some experts recommend not exceeding 50 mg twice daily (Ref). ER formulation should be avoided.

Peritoneal dialysis: Dialyzability unknown (Davison 2014):

Immediate release: Lower initial doses and an extended dosing interval (eg, 25 mg twice daily) are recommended (Ref); titrate to response. Although a maximum daily dose of 200 mg/day has been suggested (Ref), some experts recommend not exceeding 100 mg/day since a uremic state may lower the seizure threshold (Ref). ER formulation should be avoided.

Dosing: Hepatic Impairment: Adult

The hepatic dosing recommendations are based up on the best available evidence and clinical expertise. Senior Editorial Team: Matt Harris, PharmD, MHS, BCPS, FAST; Jeong Park, PharmD, MS, BCPS, FCCP, FAST; Arun Jesudian, MD; Sasan Sakiani, MD.

Note: Tramadol is a prodrug that requires conversion to the primary active metabolite (O-desmethyl tramadol) in the liver. In patients with hepatic impairment, tramadol is subject to reduced hepatic metabolism, which may result in ineffective pain control (Ref). Dose recommendations for hepatic impairment are based on the usual recommended dose of 50 to 100 mg every 4 to 6 hours for the treatment of acute and chronic pain. Use of the IR formulation is preferred over the ER formulation in patients with hepatic impairment (Ref).

Initial or dose titration in patients with preexisting liver cirrhosis :

Pain management: Moderate to severe:

Acute pain: Immediate release:

Child-Turcotte-Pugh class A: Oral: Initial: 50 mg every 8 hours as needed; may increase to 50 mg every 6 hours based on tolerability and response (maximum: 200 mg/day) (Ref).

Child-Turcotte-Pugh class B: Oral: Initial: 25 mg every 8 to 12 hours as needed; may increase to max 100 mg/day in 2 to 3 divided doses based on tolerability and response (Ref).

Child-Turcotte-Pugh class C: Use is not recommended (Ref).

Chronic pain:

Child-Turcotte-Pugh class A and B: Use of alternative analgesics (opioid or nonopioid) are preferred due to potential decreased tramadol efficacy (Ref).

Child-Turcotte-Pugh class C: Use is not recommended (Ref).

Dosage adjustment in patients with chronic, worsening hepatic function during treatment (eg, progression from Child-Turcotte-Pugh class A to B):

Pain management: Moderate to severe:

New Child-Turcotte-Pugh class A and B: Use of alternative analgesics (opioid or nonopioid) are preferred due to potential decreased tramadol efficacy (Ref).

Progression to Child-Turcotte-Pugh class C: Use is not recommended; use of alternative analgesics (opioid or nonopioid) are preferred due to potential decreased tramadol efficacy (Ref).

Acute worsening of hepatic function (eg, requiring hospitalization): Discontinue tramadol during the acute event; may resume once the acute event has resolved (ie, LFTs have stabilized or returned to baseline) (Ref).

Dosing: Pediatric

(For additional information see "Tramadol: Pediatric drug information")

Note: Doses should be titrated to appropriate analgesic effect; use the lowest effective dose for the shortest period of time:

Pain management, moderate to severe pain

Pain management, moderate to severe pain (excluding postoperative tonsillectomy/adenoidectomy pain): Note: The FDA has recommended that tramadol not be used in pediatric patients <12 years of age and all pediatric patients undergoing tonsillectomy and/or adenoidectomy due to increased risk of breathing problems (sometimes fatal). Slowed or difficult breathing has been reported in pediatric patients <18 years of age; risk may be increased in pediatric patients who are obese or have conditions such as obstructive sleep apnea or severe lung disease, or who are ultrarapid metabolizers of the drug (Ref).

Acute pain: Immediate-release formulations:

Children and Adolescents 4 to ≤16 years: Limited data available: Oral: 1 to 2 mg/kg/dose every 4 to 6 hours; maximum single dose: 100 mg (usual adult starting dose: 50 to 100 mg); maximum daily dose is the lesser of 8 mg/kg/day or 400 mg/day (Ref). Note: Due to potential respiratory complications, tramadol should be avoided in patients <12 years of age and all pediatric patients undergoing tonsillectomy and/or adenoidectomy (Ref).

Adolescents ≥17 years: Oral: 50 to 100 mg every 4 to 6 hours; maximum daily dose: 400 mg/day. For patients not requiring rapid onset of effect, tolerability to adverse effects may be improved by initiating therapy at 25 mg/day and titrating dose by 25 mg every 3 days until 25 mg 4 times daily is reached. Dose may then be increased by 50 mg every 3 days as tolerated to reach 50 mg 4 times daily.

Chronic pain: Extended-release formulations: Adolescents ≥18 years: Oral: Note: For patients requiring around-the-clock pain management for an extended period of time. Opioids, including tramadol, are not the preferred therapy for chronic noncancer pain due to insufficient evidence of benefit and risk of serious harm; nonpharmacologic treatment and nonopioid analgesics are preferred with the exception of pain from sickle cell disease and in end-of-life care (Ref). Opioids, including tramadol, should only be considered in patients who experience clinically meaningful improvement in pain and function that outweighs patient safety risks (Ref).

Patients not currently on immediate-release tramadol: 100 mg once daily; titrate every 5 days; maximum daily dose: 300 mg/day.

Patients currently on immediate-release tramadol: Calculate 24-hour total immediate-release tramadol dose and initiate total extended-release daily dose (round dose to the next lowest 100 mg increment) once daily; titrate as tolerated to desired effect; maximum daily dose: 300 mg/day.

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

Immediate release: Adolescents ≥17 years:

CrCl ≥30 mL/minute: There are no dosage adjustments provided in the manufacturer's labeling; use with caution.

CrCl <30 mL/minute: Increase dosing interval to every 12 hours; maximum daily dose: 200 mg/day.

Dialysis: Dialyzable (7%); increase dosing interval to every 12 hours; maximum daily dose: 200 mg/day; administer regular dose on the day of dialysis.

Extended release: Adolescents ≥18 years:

CrCl ≥30 mL/minute: There are no dosage adjustments provided in the manufacturer's labeling; use with caution.

CrCl <30 mL/minute: Avoid use.

Dosing: Hepatic Impairment: Pediatric

Immediate release: Adolescents ≥17 years: There are no dosage adjustments provided in the manufacturer's labeling. In patients with cirrhosis, recommended dose is 50 mg every 12 hours.

Extended release: Adolescents ≥18 years:

Mild to moderate impairment (Child-Pugh class A and B): There are no dosage adjustments provided in the manufacturer's labeling; use with caution.

Severe impairment (Child-Pugh class C): Avoid use.

Dosing: Older Adult

Elderly >65 years to ≤75 years: Refer to adult dosing; use with caution and initiate at the low end of the dosing range.

Elderly >75 years:

Immediate release: Maximum: 300 mg/day.

Extended release: Use with extreme caution.

Dosage Forms: US

Excipient information presented when available (limited, particularly for generics); consult specific product labeling. [DSC] = Discontinued product

Capsule Extended Release 24 Hour, Oral, as hydrochloride:

ConZip: 100 mg, 200 mg, 300 mg [contains fd&c blue #2 (indigo carm) aluminum lake, quinoline (d&c yellow #10) aluminum lake]

Generic: 100 mg, 150 mg [DSC], 200 mg, 300 mg

Solution, Oral, as hydrochloride:

Qdolo: 5 mg/mL (473 mL) [contains propylene glycol, sodium benzoate; grape flavor]

Generic: 5 mg/mL (5 mL)

Tablet, Oral, as hydrochloride:

Ultram: 50 mg [DSC] [scored; contains corn starch]

Generic: 50 mg, 100 mg

Tablet Extended Release 24 Hour, Oral, as hydrochloride:

Generic: 100 mg, 200 mg, 300 mg

Generic Equivalent Available: US

Yes

Dosage Forms Considerations

ConZip extended release capsules are formulated as a biphasic product, providing immediate and extended release components:

100 mg: 25 mg (immediate release) and 75 mg (extended release)

200 mg: 50 mg (immediate release) and 150 mg (extended release)

300 mg: 50 mg (immediate release) and 250 mg (extended release)

EnovaRX-Tramadol and Active-Tramadol creams are compounded from kits. Refer to manufacturer’s labeling for compounding instructions.

Synapryn FusePaq is a compounding kit for the preparation of an oral suspension. Refer to manufacturer's labeling for compounding instructions.

Dosage Forms: Canada

Excipient information presented when available (limited, particularly for generics); consult specific product labeling. [DSC] = Discontinued product

Capsule Extended Release 24 Hour, Oral, as hydrochloride:

Durela: 100 mg, 200 mg, 300 mg [contains fd&c blue #2 (indigo carm) aluminum lake, quinoline (d&c yellow #10) aluminum lake]

Tablet, Oral, as hydrochloride:

Ultram: 50 mg [DSC]

Generic: 50 mg

Tablet Extended Release 24 Hour, Oral:

Zytram XL: 75 mg [contains fd&c blue #2 (indigotine)]

Zytram XL: 150 mg, 400 mg

Tablet Extended Release 24 Hour, Oral, as hydrochloride:

Ralivia: 100 mg, 200 mg, 300 mg

Tridural: 100 mg, 200 mg, 300 mg

Zytram XL: 100 mg, 200 mg, 300 mg

Generic: 100 mg, 200 mg, 300 mg

Controlled Substance

C-IV

Medication Guide and/or Vaccine Information Statement (VIS)

An FDA-approved patient medication guide, which is available with the product information and as follows, must be dispensed with this medication:

ConZip: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/022370s020lbl.pdf#page=35

Qdolo: https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/214044s000lbl.pdf#page=27

Ultram: https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/020281s048lbl.pdf#page=42

Ultram ER: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021692s015lbl.pdf#page=36

Administration: Adult

Oral:

Immediate release: Administer without regard to meals. Measure oral solution with a calibrated oral syringe or other oral dosing device with metric units of measure (do not use household teaspoons or tablespoons); ensure accurate dosing between mg and mL.

Extended release: Swallow whole; do not crush, chew, dissolve, or split.

Capsule: Administer without regard to meals.

Tablet: Administer without regard to meals but administer in a consistent manner of either with or without meals.

Canadian products:

Durela, Ralivia, Zytram XL: Administer without regard to meals.

Tridural: Administer once daily with breakfast.

Bariatric surgery:

Capsule and tablet, extended release: Some institutions may have specific protocols that conflict with these recommendations; refer to institutional protocols as appropriate. ER capsule should be swallowed whole. Do not crush, chew, dissolve, or split as this may result in rapid release and a potentially fatal dose of tramadol. IR tablet formulation is available. If safety and efficacy can be effectively monitored, no change in formulation or administration is required after bariatric surgery; however, clinicians are advised to monitor closely for adverse effects and withdrawal symptoms after bariatric surgery. Oral morphine has been shown to have significantly increased Cmax and decreased Tmax in the immediate period (1 to 2 weeks) and long-term (6 months) period after bariatric surgery.

Administration: Pediatric

Oral:

Immediate-release tablet: May administer with or without food, but it is recommended that it be administered in a consistent manner.

Extended-release tablet: Swallow whole with a sufficient amount of liquid. Do not crush, cut, dissolve, or chew extended-release tablet; may be taken without regard to meals; tablet should be taken once daily at approximately the same time each day.

Use: Labeled Indications

Pain management:

Extended release: Management of pain severe enough to require daily, around-the-clock, long-term opioid treatment and for which alternative treatment options are inadequate.

Immediate release: Management of pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate.

Limitations of use: Reserve tramadol for use in patients for whom alternative treatment options (eg, nonopioid analgesics) are ineffective, not tolerated, or would be otherwise inadequate to provide sufficient management of pain. Tramadol ER is not indicated as an as-needed analgesic.

Use: Off-Label: Adult

Premature ejaculation

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

TraMADol may be confused with tapentadol, Toradol Trandate, traZODone, Voltaren

Ryzolt may be confused with Rydapt

Ultram may be confused with lithium, Ultane, Ultracet, Voltaren

High alert medication:

The Institute for Safe Medication Practices (ISMP) includes this medication among its list of drug classes which have a heightened risk of causing significant harm when used in error.

Older Adult: High-Risk Medication:

Beers Criteria: Tramadol is identified in the Beers Criteria as a potentially inappropriate medication to be used with caution in patients 65 years and older due to the potential to cause or exacerbate syndrome of inappropriate antidiuretic hormone secretion (SIADH) or hyponatremia; monitor sodium concentration closely when initiating or adjusting the dose in older adults (Beers Criteria [AGS 2019]).

Pediatric patients: High-risk medication:

KIDs List: Tramadol, when used in pediatric patients <18 years of age, is identified on the Key Potentially Inappropriate Drugs in Pediatrics (KIDs) list and should be used with caution due to risk of respiratory depression unless pharmacogenetic testing completed (weak recommendation; low quality of evidence) (PPA [Meyers 2020]).

International issues:

Theradol [Netherlands] may be confused with Foradil brand name for formoterol [US, Canada, and multiple international markets], Terazol brand name for terconazole [US and Canada], and Toradol brand name for ketorolac [Canada and multiple international markets]

Trexol [Mexico] may be confused with Trexall brand name for methotrexate [US]; Truxal brand name for chlorprothixene [multiple international markets]

Adverse Reactions (Significant): Considerations
CNS effects

Various CNS effects have been reported in association with tramadol use in clinical trials and case reports. In clinical trials, the most common CNS effects include dizziness, sedated state, drowsiness, headache, and central nervous system stimulation. Less commonly reported CNS adverse effects include euphoria, anxiety, depression, anger, hostility, aggression, lack of concentration, and cognitive dysfunction (Ref). In contrast, improvements in anxiety and depression symptoms have been reported during tramadol use (Ref). Euphoric effects and possible antianxiety and antidepressant effects may contribute to the addiction potential of tramadol (Ref).

Mechanism: Dose-related; tramadol metabolism to active metabolite, M1, may contribute to euphoric effect (Ref).

Risk factors:

• Dose; generally greater with higher doses

• Concurrent use of CNS depressant medications

• CYP2D6 ultra-rapid metabolizers

Constipation

Constipation has been reported in patients taking opioids, including tramadol (Ref).

Mechanism: Dose-related; related to the pharmacologic action (ie, activation of mu opioid receptors in the GI tract resulting in decreased peristalsis, reduced mucosal secretions, and delayed gastric emptying) (Ref).

Onset: Intermediate; occurs within first 4 weeks of therapy (Ref). Minimal to no tolerance to constipation develops with chronic use ((Ref).

Risk factors:

• Dose; generally greater with higher doses

Hyponatremia

Hyponatremia has been reported with tramadol use; many cases have been severe (sodium <120 mmol/L). Some cases have occurred due to the syndrome of inappropriate antidiuretic hormone.

Onset: Varied; most cases have occurred within the first week of initiation.

Risk factors:

• Females >65 years of age may be at higher risk

Respiratory depression

Life-threatening or fatal respiratory depression has occurred with opioids, including tramadol, at therapeutic and supratherapeutic doses in adult and pediatric patients (Ref). Respiratory depression may be reversible with medical intervention (Ref). Tramadol may be associated with less risk of respiratory depression than other opioids in adults (Ref); in pediatric patients, a serious (sometimes fatal) risk of respiratory depression exists with tramadol and is similar to that of opioids such as codeine (Ref).

Mechanism: Dose-related; related to the pharmacologic action (ie, activation of mu opioid receptors in the CNS respiratory center) (Ref).

Onset: Rapid; mean of 8 hours after ingestion (range: 1 to 24 hours); dependent on dose and patient variables (eg, CYP2D6 ultra-rapid metabolizer, kidney impairment) (Ref).

Risk factors:

• Dose; generally greater with higher doses (Ref)

• Age ≥65 years (Ref)

• Cachexia

• Cardiovascular disease (Ref)

• Chronic pulmonary disease (Ref)

• Concurrent use of other psychoactive medications (eg, benzodiazepines or CNS depressants, antipsychotics) (Ref)

• CYP2D6 ultra-rapid metabolizers (adult and pediatric) (Ref)

• Debilitation

• Depression (or other concurrent psychiatric illness) (Ref)

• Kidney or liver impairment (Ref)

• Substance use disorder (Ref)

• Pediatric:

• Age <12 years of age (Ref)

• Age ≥12 years with comorbid conditions like obesity, obstructive sleep apnea, or severe lung disease, which may increase the risk of serious breathing problem (Ref)

Seizures

Seizure has been reported after therapeutic and supratherapeutic doses of tramadol (Ref). Seizures typically present as a single generalized tonic-clonic episode lasting <5 minutes (Ref); although, recurrent seizures have been reported (Ref).

Mechanism: Dose-related. Mechanism not fully elucidated; may be related to excessive serotonergic activity (Ref), opioid-dependent GABA receptor inhibition (Ref), and/or opioid-receptor dependent histamine release (Ref).

Onset: Rapid; occurs within 4 to 6 hours of ingestion (Ref).

Risk factors:

• Dose; generally greater with higher doses (Ref)

• Concurrent medications: Selective serotonin reuptake inhibitors (SSRIs), serotonin norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs) and other tricyclic compounds (eg, cyclobenzaprine, promethazine), other opioids, monoamine oxidase inhibitors (MAOIs), anorectics, neuroleptics, drugs that reduce the seizure threshold, and/or drugs that impair metabolism of tramadol (eg, CYP2D6 and 3A4 inhibitors)

• CYP2D6 poor metabolizers (Ref)

• History of seizure disorder

• Patients otherwise at risk for seizures (ie, alcohol withdrawal, CNS infections, head trauma)

Serotonin syndrome

Life-threatening serotonin syndrome has been reported after therapeutic and supratherapeutic doses of tramadol (Ref). Risk is increased when tramadol is administered in combination with agents that increase its plasma concentrations (eg, CYP2D6 and CYP3A4 inhibitors) and with other serotonergic agents. However, it has been reported after tramadol monotherapy; more often observed in the setting of overdose (Ref). Serotonin syndrome may be reversible with medical intervention (Ref).

Mechanism: Excessive serotonin concentrations at the synaptic cleft and/or inhibition of tramadol metabolism leading to increased tramadol concentrations (Ref).

Onset: Varied; may occur within several hours to days after administration or may present later.

Risk factors:

• Concurrent use of serotonergic medications: Selective serotonin reuptake inhibitors (SSRIs), serotonin norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), monoamine oxidase inhibitors (MAOIs), triptans, etc. (Ref)

• Concurrent use of CYP2D6 and/or CYP3A4 inhibitors (Ref)

• CYP2D6 poor metabolizers (Ref)

• Age >65 years (Ref)

Withdrawal syndrome

Physical dependence, manifesting as a withdrawal syndrome, has been observed after abrupt discontinuation of tramadol (Ref). Reported symptoms are consistent with both opioid and serotonin withdrawal (Ref). Withdrawal symptoms may occur after several days or longer therapy durations. Withdrawal symptoms usually resolve within 2 to 7 days (Ref).

Mechanism: Withdrawal; autonomic hyperexcitability in the absence of opioid agonist suppressive effects (Ref); reduced availability of serotonin (Ref).

Onset: Rapid; symptom onset typically occurs within 24 hours of discontinuing tramadol therapy and usually resolves within 2 to 7 days (Ref).

Risk factors:

• Abrupt discontinuation of tramadol in physically dependent patients

• Concurrent use of opioid mixed agonist/antagonists, opioid partial agonists, or naloxone (Ref)

• Duration of use (potential risk factor) (Ref)

• Higher doses (Ref)

Adverse Reactions

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

>10%:

Gastrointestinal: Constipation (9% to 21%; placebo: 4%), dyspepsia, nausea (15% to 26%), xerostomia (5% to 13%)

Nervous system: Dizziness (10% to 23%; placebo: 5% to 7%), drowsiness (7% to 16%; placebo: 2% to 4%), headache (12% to 23%; placebo: 11% to 20%), vertigo

1% to 10%:

Cardiovascular: Chest pain (1% to <5%), flushing (8% to 10%), hypertension (1% to <5%), orthostatic hypotension (≤4%), peripheral edema (<5%), vasodilation (1% to <5%)

Dermatologic: Dermatitis (1% to <5%), diaphoresis (2% to 7%), pruritus (3% to 9%), skin rash (1% to <5%)

Endocrine & metabolic: Hot flash (1% to <5%), hyperglycemia (1% to <5%), weight loss (1% to <5%)

Gastrointestinal: Abdominal pain (1% to <5%; upper abdominal pain: 1% to <5%), anorexia (1% to 6%), decreased appetite (1% to <5%), diarrhea (7% to 9%), flatulence (<5%), vomiting (5% to 10%)

Genitourinary: Menopausal symptoms (1% to <5%), pelvic pain (1% to <5%), prostatic disease (1% to <5%), urinary frequency (<5%), urinary retention (<5%), urinary tract infection (1% to <5%)

Nervous system: Agitation (<5%), anxiety (1% to <5%), apathy (1% to <5%), ataxia (1% to <5%), chills (1% to <5%), confusion (1% to <5%), depersonalization (1% to <5%), depression (1% to <5%), euphoria (<5%), falling (1% to <5%), hypertonia (<5%), hypoesthesia (1% to <5%), insomnia (5% to 9%), lethargy (1% to <5%), malaise (<5%), nervousness (1% to <5%), paresthesia (1% to <5%), restlessness (1% to <5%), rigors (1% to <5%), sleep disorder (<5%), withdrawal syndrome (<5%)

Neuromuscular & skeletal: Arthralgia (1% to <5%), asthenia (4% to 9%), back pain (1% to <5%), increased creatine phosphokinase in blood specimen (1% to <5%), limb pain (1% to <5%), myalgia (<5%), neck pain (1% to <5%), tremor (<5%)

Ophthalmic: Blurred vision (1% to <5%), miosis (1% to <5%)

Respiratory: Bronchitis (1% to <5%), cough (1% to <5%), dyspnea (1% to <5%), flu-like symptoms (1% to <5%), nasal congestion (1% to <5%), nasopharyngitis (1% to <5%), pharyngitis (1% to <5%), rhinitis (1% to <5%), rhinorrhea (1% to <5%), sinusitis (1% to <5%), sneezing (1% to <5%), upper respiratory tract infection (1% to <5%)

Miscellaneous: Accidental injury (<5%), fever (1% to <5%)

<1%:

Cardiovascular: Acute myocardial infarction, hypotension, ischemic heart disease, lower extremity edema, palpitations, peripheral ischemia, syncope, tachycardia

Dermatologic: Cellulitis, cold and clammy skin, ecchymoses, night sweats, piloerection, skin vesicle, Stevens-Johnson syndrome (Mockenhaupt 2008), toxic epidermal necrolysis (Mockenhaupt 2008), urticaria

Endocrine & metabolic: Decreased libido, gout, increased gamma-glutamyl transferase, menstrual disease

Gastrointestinal: Appendicitis, cholecystitis, cholelithiasis, dysgeusia, gastroenteritis, pancreatitis

Genitourinary: Cystitis, dysuria, hematuria

Hematologic & oncologic: Anemia, bruise

Nervous system: Abnormal dreams, cognitive dysfunction, disorientation, emotional lability, hallucination, irritability, lack of concentration, migraine, sedated state, seizure (Memarian 2018), serotonin syndrome (Shakoor 2014), suicidal tendencies, yawning

Neuromuscular & skeletal: Hyperkinetic muscle activity, joint stiffness, lower limb cramp, muscle cramps, muscle spasm, muscle twitching, neck stiffness

Otic: Otitis, tinnitus

Respiratory: Pneumonia

Frequency not defined:

Hypersensitivity: Angioedema

Neuromuscular & skeletal: Muscle spasticity

Respiratory: Bronchospasm

Postmarketing:

Cardiovascular: Prolonged QT interval on ECG, pulmonary embolism, torsades de pointes

Dermatologic: Erythema multiforme (Sanchez-Gonzalez 2020)

Endocrine & metabolic: Adrenocortical insufficiency (Debono 2011), hypoglycemia (within 30 days of initiation [Fournier 2015, Odonkor 2016]; may occur more often in patients with predisposing risk factors [eg, diabetes] and may result in hospitalization), hyponatremia

Gastrointestinal: Gastrointestinal hemorrhage, stomatitis

Genitourinary: Proteinuria

Hepatic: Hepatic failure, hepatitis

Hypersensitivity: Anaphylaxis (<0.1%) (Mori 2015)

Nervous system: Delirium (Agrawal 2009, Kunig 2006)

Neuromuscular & skeletal: Femoral neck fracture (Wei 2020)

Ophthalmic: Cataract, mydriasis (Makris 2012)

Otic: Deafness

Respiratory: Pulmonary edema, respiratory depression (Hassanian-Moghaddam 2013)

Contraindications

Hypersensitivity (eg, anaphylaxis) to tramadol, opioids, or any component of the formulation; pediatric patients <12 years of age; postoperative management in pediatric patients <18 years of age who have undergone tonsillectomy and/or adenoidectomy; significant respiratory depression; acute or severe bronchial asthma in the absence of appropriately monitored settings and/or resuscitative equipment; GI obstruction, including paralytic ileus (known or suspected); concomitant use with or within 14 days following monoamine oxidase inhibitor therapy.

Canadian products: Additional contraindications (not in US labeling): (Note: Contraindications may differ between product labeling; refer also to product labeling): Severe renal impairment (CrCl <30 mL/minute), severe hepatic impairment (Child-Pugh class C); mild, intermittent, or short-duration pain that can be managed with other pain medication; management of perioperative pain; status asthmaticus, chronic obstructive airway, acute respiratory depression, hypercapnia, cor pulmonale, delirium tremens, seizure disorder, severe CNS depression, increased cerebrospinal or intracranial pressure, brain tumor, head injury, suspected surgical abdomen (eg, acute appendicitis or pancreatitis); acute intoxication with ethanol, hypnotics, centrally acting analgesics, opioids, or psychotropic drugs; breastfeeding, pregnancy; use during labor and delivery.

Warnings/Precautions

Concerns related to adverse effects:

• Hypotension: May cause severe hypotension (including orthostatic hypotension and syncope); use with caution in patients with hypovolemia, cardiovascular disease (including acute myocardial infarction [MI]), or drugs that may exaggerate hypotensive effects (including phenothiazines or general anesthetics). Monitor for symptoms of hypotension following initiation or dose titration. Avoid use in patients with circulatory shock.

Disease-related concerns:

• Abdominal conditions: May obscure diagnosis or clinical course of patients with acute abdominal conditions.

• Adrenocortical insufficiency: Use with caution in patients with adrenal insufficiency, including Addison disease. Long-term opioid use may cause secondary hypogonadism, which may lead to mood disorders and osteoporosis (Brennan 2013).

• Biliary tract impairment: Use caution in patients with biliary tract dysfunction or acute pancreatitis; opioids may cause spasm of the sphincter of Oddi.

• CNS depression/coma: Avoid use in patients with impaired consciousness or coma as these patients are susceptible to intracranial effects of CO2 retention.

• Diabetes: Use with caution in patients with diabetes; tramadol may cause hypoglycemia.

• Delirium tremens: Use with caution in patients with delirium tremens.

• Head trauma: Use with extreme caution in patients with head injury, intracranial lesions, or elevated intracranial pressure (ICP); exaggerated elevation of ICP may occur.

• Hepatic impairment: Use with caution; dosage adjustments may be required. ER formulations should not be used in severe hepatic impairment (Child-Pugh class C).

• Mental health conditions: Use opioids with caution for chronic pain in patients with mental health conditions (eg, depression, anxiety disorders, post-traumatic stress disorder) due to increased risk for opioid use disorder and overdose; more frequent monitoring is recommended (CDC [Dowell 2016]).

• Obesity: Use with caution in patients who are morbidly obese.

• Prostatic hyperplasia/urinary stricture: Use with caution in patients with prostatic hyperplasia and/or urinary stricture.

• Psychosis: Use with caution in patients with toxic psychosis.

• Renal impairment: Use with caution; reduce dosage of IR formulations in patients with severe renal impairment; ER formulations should be avoided in severe renal impairment.

• Respiratory disease: Use with caution and monitor for respiratory depression in patients with significant chronic obstructive pulmonary disease or cor pulmonale, and those with a substantially decreased respiratory reserve, hypoxia, hypercapnia, or preexisting respiratory depression, particularly when initiating and titrating therapy; critical respiratory depression may occur, even at therapeutic dosages. Consider the use of alternative nonopioid analgesics in these patients.

• Sleep-related disorders: Opioid use increases the risk for sleep-related disorders (eg, central sleep apnea [CSA], hypoxemia) in a dose-dependent fashion. Use with caution for chronic pain and titrate dosage cautiously in patients with risk factors for sleep-disordered breathing (eg, heart failure, obesity). Consider dose reduction in patients presenting with CSA. Avoid opioids in patients with moderate to severe sleep-disordered breathing (CDC [Dowell 2016]).

• Suicide risk: Avoid use in patients who are suicidal; use with caution in patients taking tranquilizers and/or antidepressants, or those with an emotional disturbance including depression. Consider the use of alternative nonopioid analgesics in these patients.

• Thyroid dysfunction: Use with caution in patients with thyroid dysfunction.

Special populations:

• CYP2D6 "poor metabolizers": Poor metabolizers have decreased metabolism of tramadol to its active metabolite, which may diminish analgesia; avoid the use of tramadol and consider alternatives that are not metabolized by CYP2D6 (CPIC [Crews 2021]).

• CYP2D6 "ultrarapid metabolizers": Ultrarapid metabolizers have increased metabolism of tramadol to its active metabolite, which may increase the risk of toxicity; avoid the use of tramadol and consider alternatives that are not metabolized by CYP2D6 (CPIC [Crews 2021]). The occurrence of this phenotype is seen in ~1% to 2% of East Asian patients (Chinese, Japanese, Korean), 1% to 10% of Caucasian patients, 3% to 4% of Black patients, and may be >10% in certain racial/ethnic groups (ie, Oceanian, Northern African, Middle Eastern, Ashkenazi Jewish, and Puerto Rican patients). Deaths have also occurred in breastfeeding infants after being exposed to high concentrations of morphine because the mothers were ultrarapid metabolizers of codeine.

• Older adult: Use opioids for chronic pain with caution in older adults; monitor closely due to an increased potential for risks, including certain risks such as falls/fracture, cognitive impairment, and constipation. Clearance may also be reduced in older adults (with or without renal impairment) resulting in a narrow therapeutic window and increasing the risk for respiratory depression or overdose (CDC [Dowell 2016]). Consider the use of alternative nonopioid analgesics in these patients.

• Pediatric: Respiratory depression:Risk factors include conditions associated with hypoventilation, such as postoperative status, obstructive sleep apnea, obesity, severe pulmonary disease, neuromuscular disease, and concomitant use of other medications that cause respiratory depression.

Other warnings/precautions:

• Abuse/misuse/diversion: Use with caution in patients with a history of drug abuse or acute alcoholism; potential for drug dependency exists. Other risk factors associated with increased risk include a personal or family history of substance use disorder or mental illness (eg, major depression).

• Appropriate use: Chronic pain (outside of end-of-life or palliative care, active cancer treatment, sickle cell disease, or medication-based opioid use disorder treatment) in outpatient setting in adults: Opioids should not be used as first-line therapy for chronic pain management (pain >3-month duration or beyond time of normal tissue healing) due to limited short-term benefits, undetermined long-term benefits, and association with serious risks (eg, overdose, MI, auto accidents, risk of developing opioid use disorder). Preferred management includes nonpharmacologic therapy and nonopioid therapy (eg, nonsteroidal anti-inflammatory drugs, acetaminophen, certain antiseizure and antidepressant medications). If opioid therapy is initiated, it should be combined with nonpharmacologic and nonopioid therapy, as appropriate. Prior to initiation, known risks of opioid therapy should be discussed and realistic treatment goals for pain/function should be established, including consideration for discontinuation if benefits do not outweigh risks. Therapy should be continued only if clinically meaningful improvement in pain/function outweighs risks. Therapy should be initiated at the lowest effective dosage using IR opioids (instead of ER/long-acting opioids). Risk associated with use increases with higher opioid dosages. Risks and benefits should be re-evaluated when increasing dosage to ≥50 MME/day orally; dosages ≥90 MME/day orally should be avoided unless carefully justified (CDC [Dowell 2016]).

• Naloxone access: Discuss the availability of naloxone with all patients who are prescribed opioid analgesics, as well as their caregivers, and consider prescribing it to patients who are at increased risk of opioid overdose. These include patients who are also taking benzodiazepines or other CNS depressants, have an opioid use disorder (OUD) (current or history of), have experienced a previous opioid overdose, have a history of a substance use disorder, or have higher opioid dosages (≥50 morphine milligram equivalents [MME]/day orally) (CDC [Dowell 2016]). Additionally, health care providers should consider prescribing naloxone to patients prescribed medications to treat OUD; patients at risk of opioid overdose even if they are not taking an opioid analgesic or medication to treat OUD; and patients taking opioids, including methadone or buprenorphine for OUD, if they have household members, including children, or other close contacts at risk for accidental ingestion or opioid overdose. Inform patients and caregivers on options for obtaining naloxone (eg, by prescription, directly from a pharmacist, a community-based program) as permitted by state dispensing and prescribing guidelines. Educate patients and caregivers on how to recognize respiratory depression, proper administration of naloxone, and getting emergency help.

• Optimal regimen: An opioid-containing analgesic regimen should be tailored to each patient's needs and based upon the type of pain being treated (acute versus chronic), the route of administration, degree of tolerance for opioids (naive versus chronic user), age, weight, and medical condition. The optimal analgesic dose varies widely among patients; doses should be titrated to pain relief/prevention.

• Surgery: Opioids decrease bowel motility; monitor for decrease bowel motility in postop patients receiving opioids. Use with caution in the perioperative setting; individualize treatment when transitioning from parenteral to oral analgesics.

Warnings: Additional Pediatric Considerations

In April 2017, the FDA announced tramadol use should be avoided in all pediatric patients <12 years and all pediatric patients undergoing tonsillectomy or adenoidectomy. The FDA is requiring updated manufacturer labeling to include in the following contraindications: Use in patients <12 years to treat pain and use in patients <18 years to treat postoperative tonsillectomy/adenoidectomy pain (FDA 2017).

Metabolism/Transport Effects

Substrate of CYP2B6 (minor), CYP2D6 (major), 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.

Ajmaline: May increase the serum concentration of CYP2D6 Substrates (High risk with Inhibitors). Risk C: Monitor therapy

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

Almotriptan: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Alvimopan: Opioid Agonists may enhance the adverse/toxic effect of Alvimopan. This is most notable for patients receiving long-term (i.e., more than 7 days) opiates prior to alvimopan initiation. Management: Alvimopan is contraindicated in patients receiving therapeutic doses of opioids for more than 7 consecutive days immediately prior to alvimopan initiation. Risk D: Consider therapy modification

Amifampridine: Agents With Seizure Threshold Lowering Potential may enhance the neuroexcitatory and/or seizure-potentiating effect of Amifampridine. Risk C: Monitor therapy

Amphetamines: May enhance the analgesic effect of Opioid Agonists. Risk C: Monitor therapy

Amphetamines: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability). Initiate amphetamines at lower doses, monitor frequently, and adjust doses as needed. Risk C: Monitor therapy

Androgens: May enhance the hypoglycemic effect of Agents with Blood Glucose Lowering Effects. Risk C: Monitor therapy

Anticholinergic Agents: May enhance the adverse/toxic effect of Opioid Agonists. Specifically, the risk for constipation and urinary retention may be increased with this combination. Risk C: Monitor therapy

Antidiabetic Agents: May enhance the hypoglycemic effect of Hypoglycemia-Associated Agents. Risk C: Monitor therapy

Antiemetics (5HT3 Antagonists): May enhance the serotonergic effect of TraMADol. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Artemether and Lumefantrine: May increase the serum concentration of CYP2D6 Substrates (High risk with Inhibitors). 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

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

BuPROPion: May enhance the neuroexcitatory and/or seizure-potentiating effect of Agents With Seizure Threshold Lowering Potential. Risk C: Monitor therapy

BusPIRone: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

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

CarBAMazepine: TraMADol may enhance the CNS depressant effect of CarBAMazepine. TraMADol may diminish the therapeutic effect of CarBAMazepine. CarBAMazepine may decrease the serum concentration of TraMADol. Risk X: Avoid combination

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

Clofazimine: May increase the serum concentration of CYP3A4 Substrates (High risk with Inhibitors). Risk C: Monitor therapy

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

CYP2D6 Inhibitors (Moderate): May decrease serum concentrations of the active metabolite(s) of TraMADol. CYP2D6 Inhibitors (Moderate) may increase the serum concentration of TraMADol. Risk C: Monitor therapy

CYP2D6 Inhibitors (Strong): May decrease serum concentrations of the active metabolite(s) of TraMADol. CYP2D6 Inhibitors (Strong) may increase the serum concentration of TraMADol. Risk C: Monitor therapy

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

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

CYP3A4 Inhibitors (Moderate): May increase serum concentrations of the active metabolite(s) of TraMADol. CYP3A4 Inhibitors (Moderate) may increase the serum concentration of TraMADol. Risk C: Monitor therapy

CYP3A4 Inhibitors (Strong): May increase serum concentrations of the active metabolite(s) of TraMADol. CYP3A4 Inhibitors (Strong) may increase the serum concentration of TraMADol. Risk C: Monitor therapy

Dapoxetine: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Do not use serotonergic agents (high risk) with dapoxetine or within 7 days of serotonergic agent discontinuation. Do not use dapoxetine within 14 days of monoamine oxidase inhibitor use. Dapoxetine labeling lists this combination as contraindicated. Risk X: Avoid combination

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

Desmopressin: Opioid Agonists may enhance the hyponatremic effect of Desmopressin. Risk C: Monitor therapy

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

Dexmethylphenidate-Methylphenidate: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Dextromethorphan: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

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

Digoxin: TraMADol may increase the serum concentration of Digoxin. Risk C: Monitor therapy

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

Diuretics: Opioid Agonists may enhance the adverse/toxic effect of Diuretics. Opioid Agonists may diminish the therapeutic effect of Diuretics. 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

DULoxetine: May enhance the adverse/toxic effect of TraMADol. The risk for serotonin syndrome/serotonin toxicity and seizures may be increased with this combination. DULoxetine may diminish the therapeutic effect of TraMADol. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes), reduced tramadol effectiveness and seizures if these agents are combined. Risk C: Monitor therapy

Eletriptan: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Eluxadoline: Opioid Agonists may enhance the constipating effect of Eluxadoline. Risk X: Avoid combination

Ergot Derivatives: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Fexinidazole: May increase the serum concentration of CYP3A4 Substrates (High risk with Inhibitors). Risk X: Avoid combination

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

Fusidic Acid (Systemic): May increase the serum concentration of CYP3A4 Substrates (High risk with Inhibitors). Risk X: Avoid combination

Gastrointestinal Agents (Prokinetic): Opioid Agonists may diminish the therapeutic effect of Gastrointestinal Agents (Prokinetic). Risk C: Monitor therapy

Herbal Products with Glucose Lowering Effects: May enhance the hypoglycemic effect of Hypoglycemia-Associated Agents. 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

Hypoglycemia-Associated Agents: May enhance the hypoglycemic effect of other Hypoglycemia-Associated Agents. Risk C: Monitor therapy

Iobenguane Radiopharmaceutical Products: TraMADol may diminish the therapeutic effect of Iobenguane Radiopharmaceutical Products. Management: Discontinue all drugs that may inhibit or interfere with catecholamine transport or uptake for at least 5 biological half-lives before iobenguane administration. Do not administer tramadol until at least 7 days after each iobenguane dose. Risk X: Avoid combination

Iohexol: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Iohexol. Specifically, the risk for seizures may be increased. Management: Discontinue agents that may lower the seizure threshold 48 hours prior to intrathecal use of iohexol. Wait at least 24 hours after the procedure to resume such agents. In nonelective procedures, consider use of prophylactic antiseizure drugs. Risk D: Consider therapy modification

Iomeprol: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Iomeprol. Specifically, the risk for seizures may be increased. Management: Discontinue agents that may lower the seizure threshold 48 hours prior to intrathecal use of iomeprol. Wait at least 24 hours after the procedure to resume such agents. In nonelective procedures, consider use of prophylactic antiseizure drugs. Risk D: Consider therapy modification

Iopamidol: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Iopamidol. Specifically, the risk for seizures may be increased. Management: Discontinue agents that may lower the seizure threshold 48 hours prior to intrathecal use of iopamidol. Wait at least 24 hours after the procedure to resume such agents. In nonelective procedures, consider use of prophylactic antiseizure drugs. 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

Linezolid: May enhance the serotonergic effect of Serotonergic Opioids (High Risk). This could result in serotonin syndrome. Management: Consider alternatives to this drug combination. If combined, monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes). 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

Lorcaserin (Withdrawn From US Market): May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

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

Maitake: May enhance the hypoglycemic effect of Agents with Blood Glucose Lowering Effects. 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

Methylene Blue: May enhance the serotonergic effect of Serotonergic Opioids (High Risk). This could result in serotonin syndrome. Management: Consider alternatives to this drug combination. If combined, monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes). Risk D: Consider therapy modification

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

Metoclopramide: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Consider monitoring for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

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

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

Monoamine Oxidase Inhibitors (Antidepressant): May enhance the adverse/toxic effect of TraMADol. Specifically, the risk for serotonin syndrome/serotonin toxicity and seizures may be increased.. Risk X: Avoid combination

Monoamine Oxidase Inhibitors (Type B): Serotonergic Opioids (High Risk) may enhance the serotonergic effect of Monoamine Oxidase Inhibitors (Type B). This could result in serotonin syndrome. Risk X: Avoid combination

Nalfurafine: Opioid Agonists may enhance the adverse/toxic effect of Nalfurafine. Opioid Agonists may diminish the therapeutic effect of Nalfurafine. Risk C: Monitor therapy

Nalmefene: May diminish the therapeutic effect of Opioid Agonists. Management: Avoid the concomitant use of oral nalmefene and opioid agonists. Discontinue oral nalmefene 1 week prior to any anticipated use of opioid agonists. If combined, larger doses of opioid agonists will likely be required. Risk D: Consider therapy modification

Naltrexone: May diminish the therapeutic effect of Opioid Agonists. Management: Seek therapeutic alternatives to opioids. See full drug interaction monograph for detailed recommendations. Risk X: Avoid combination

Nefazodone: May enhance the adverse/toxic effect of TraMADol. Specifically, the risk for serotonin syndrome/serotonin toxicity and seizures may be increased. Nefazodone may increase the serum concentration of TraMADol. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes), seizures, and tramadol adverse effects when these agents are combined. Risk C: Monitor therapy

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

Ondansetron: May enhance the serotonergic effect of TraMADol. This could result in serotonin syndrome. Ondansetron may diminish the therapeutic effect of TraMADol. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) and diminished tramadol efficacy when these agents are combined. Risk C: Monitor therapy

Opioids (Mixed Agonist / Antagonist): May diminish the analgesic effect of Opioid Agonists. Management: Seek alternatives to mixed agonist/antagonist opioids in patients receiving pure opioid agonists, and monitor for symptoms of therapeutic failure/high dose requirements (or withdrawal in opioid-dependent patients) if patients receive these combinations. Risk X: Avoid combination

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

Oxitriptan: Serotonergic Agents (High Risk) may enhance the serotonergic effect of Oxitriptan. This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

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

Ozanimod: May enhance the adverse/toxic effect of Serotonergic Agents (High Risk). Risk C: Monitor therapy

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

Peginterferon Alfa-2b: May decrease the serum concentration of CYP2D6 Substrates (High risk with Inhibitors). Peginterferon Alfa-2b may increase the serum concentration of CYP2D6 Substrates (High risk with Inhibitors). Risk C: Monitor therapy

Pegvisomant: Opioid Agonists may diminish the therapeutic effect of Pegvisomant. Risk C: Monitor therapy

Pegvisomant: May enhance the hypoglycemic effect of Agents with Blood Glucose Lowering Effects. Risk C: Monitor therapy

PHENobarbital: May enhance the CNS depressant effect of TraMADol. PHENobarbital may decrease the serum concentration of TraMADol. Management: Avoid use of tramadol and phenobarbital when possible. Monitor for respiratory depression/sedation. Because phenobarbital is also a strong CYP3A4 inducer, monitor for decreased tramadol efficacy and withdrawal if combined. Risk D: Consider therapy modification

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

Primidone: May enhance the CNS depressant effect of TraMADol. Primidone may decrease the serum concentration of TraMADol. Management: Avoid use of tramadol and primidone when possible. Monitor for respiratory depression/sedation. Because primidone is also a strong CYP3A4 inducer, monitor for decreased tramadol efficacy and withdrawal if combined. Risk D: Consider therapy modification

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

Prothionamide: May enhance the hypoglycemic effect of Agents with Blood Glucose Lowering Effects. Risk C: Monitor therapy

Quinolones: May enhance the hypoglycemic effect of Agents with Blood Glucose Lowering Effects. Quinolones may diminish the therapeutic effect of Agents with Blood Glucose Lowering Effects. Specifically, if an agent is being used to treat diabetes, loss of blood sugar control may occur with quinolone use. Risk C: Monitor therapy

Ramosetron: Opioid Agonists may enhance the constipating effect of Ramosetron. 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

Salicylates: May enhance the hypoglycemic effect of Agents with Blood Glucose Lowering Effects. Risk C: Monitor therapy

Samidorphan: May diminish the therapeutic effect of Opioid Agonists. Risk X: Avoid combination

Selective Serotonin Reuptake Inhibitors: TraMADol may enhance the adverse/toxic effect of Selective Serotonin Reuptake Inhibitors. Specifically, the risk for serotonin syndrome/serotonin toxicity and seizures may be increased. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) and seizures when these agents are combined. Risk C: Monitor therapy

Selective Serotonin Reuptake Inhibitors (Strong CYP2D6 Inhibitors): May enhance the adverse/toxic effect of TraMADol. Specifically, the risk for serotonin syndrome/serotonin toxicity and seizures may be increased. Selective Serotonin Reuptake Inhibitors (Strong CYP2D6 Inhibitors) may diminish the therapeutic effect of TraMADol. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes), seizures, and decreased tramadol efficacy when these agents are combined. Risk C: Monitor therapy

Serotonergic Agents (High Risk, Miscellaneous): Serotonergic Opioids (High Risk) may enhance the serotonergic effect of Serotonergic Agents (High Risk, Miscellaneous). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) if these agents are combined. Risk C: Monitor therapy

Serotonergic Non-Opioid CNS Depressants: May enhance the CNS depressant effect of Serotonergic Opioids (High Risk). Serotonergic Non-Opioid CNS Depressants may enhance the serotonergic effect of Serotonergic Opioids (High Risk). This could result in serotonin syndrome. Management: Consider alternatives to this drug combination. If combined, monitor for signs and symptoms of serotonin syndrome/serotonin toxicity and CNS depression. Risk D: Consider therapy modification

Serotonergic Opioids (High Risk): May enhance the CNS depressant effect of TraMADol. Serotonergic Opioids (High Risk) may enhance the serotonergic effect of TraMADol. This could result in serotonin syndrome. Management: Consider alternatives to this drug combination. If combined, monitor for signs and symptoms of serotonin syndrome/serotonin toxicity and CNS depression. Risk D: Consider therapy modification

Serotonin 5-HT1D Receptor Agonists (Triptans): May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Serotonin/Norepinephrine Reuptake Inhibitors: May enhance the adverse/toxic effect of TraMADol. Specifically, the risk for serotonin syndrome/serotonin toxicity and seizures may be increased. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) and seizures when these agents are combined. Risk C: Monitor therapy

Sincalide: Drugs that Affect Gallbladder Function may diminish the therapeutic effect of Sincalide. Management: Consider discontinuing drugs that may affect gallbladder motility prior to the use of sincalide to stimulate gallbladder contraction. Risk D: Consider therapy modification

Sodium Phosphates: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Sodium Phosphates. Specifically, the risk of seizure or loss of consciousness may be increased in patients with significant sodium phosphate-induced fluid or electrolyte abnormalities. Risk C: Monitor therapy

Somatostatin Analogs: Opioid Agonists may diminish the analgesic effect of Somatostatin Analogs. Opioid Agonists may enhance the analgesic effect of Somatostatin Analogs. Risk C: Monitor therapy

St John's Wort: May enhance the serotonergic effect of TraMADol. This could result in serotonin syndrome. St John's Wort may decrease the serum concentration of TraMADol. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity and reduced tramadol effects (including withdrawal symptoms) when combined. Monitor for increased tramadol effects if St John's wort is discontinued. Risk C: Monitor therapy

Succinylcholine: May enhance the bradycardic effect of Opioid Agonists. 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

Syrian Rue: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

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

Tricyclic Antidepressants: May enhance the CNS depressant effect of Serotonergic Opioids (High Risk). Serotonergic Opioids (High Risk) may enhance the serotonergic effect of Tricyclic Antidepressants. This could result in serotonin syndrome. Management: Consider alternatives to this drug combination. If combined, monitor for signs and symptoms of serotonin syndrome/serotonin toxicity and CNS depression. Risk D: Consider therapy modification

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

Vitamin K Antagonists (eg, warfarin): TraMADol may enhance the anticoagulant effect of Vitamin K Antagonists. 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

Reproductive Considerations

Long-term opioid use may cause secondary hypogonadism, which may lead to sexual dysfunction or infertility in men and women (Brennan 2013).

Premature ejaculation may contribute to male infertility. Tramadol may be an alternative treatment for this condition; however, due to the risk of addiction and adverse effects associated with opioid use, it should only be used in patients who have experienced treatment failure with other therapies (ISSM [Althof 2014]; Martyn-St. James 2015).

Pregnancy Considerations

Tramadol crosses the placenta.

According to some studies, maternal use of opioids may be associated with birth defects (including neural tube defects, congenital heart defects, and gastroschisis), poor fetal growth, stillbirth, and preterm delivery (CDC [Dowell 2016]).

[US Boxed Warning]: Prolonged use of tramadol during pregnancy can result in neonatal opioid withdrawal syndrome, which may be life-threatening if not recognized and treated, and requires management according to protocols developed by neonatology experts. If opioid use is required for a prolonged period in a pregnant woman, advise the patient of the risk of neonatal opioid withdrawal syndrome and ensure that appropriate treatment will be available. If chronic opioid exposure occurs in pregnancy, adverse events in the newborn (including withdrawal) may occur (Chou 2009). Symptoms of neonatal abstinence syndrome (NAS) following opioid exposure may be autonomic (eg, fever, temperature instability), gastrointestinal (eg, diarrhea, vomiting, poor feeding/weight gain), or neurologic (eg, high-pitched crying, hyperactivity, increased muscle tone, increased wakefulness/abnormal sleep pattern, irritability, sneezing, seizure, tremor, yawning) (Dow 2012; Hudak 2012). Mothers who are physically dependent on opioids may give birth to infants who are also physically dependent. Opioids may cause respiratory depression and psycho-physiologic effects in the neonate; newborns of mothers receiving opioids during labor should be monitored.

Tramadol is not commonly used to treat pain during labor and immediately postpartum (ACOG 209 2019) or chronic noncancer pain in pregnant women or those who may become pregnant (CDC [Dowell 2016]; Chou 2009).

Breastfeeding Considerations

Tramadol and the active M1 metabolite are present in breast milk. M1 has stronger opioid activity than tramadol. Actual exposure to a breastfeeding infant may depend on the mothers CYP2D6 metabolism (Salman 2011).

Tramadol is not recommended for use in breastfeeding women. Due to the potential for serious adverse events in the breastfed infant (including excess sedation and respiratory depression), use during breastfeeding is not recommended by the manufacturer. Nonopioid analgesics are preferred for breastfeeding females who require pain control peripartum or for surgery outside of the postpartum period (ABM [Martin 2018]; ABM [Reece-Stremtan 2017]). When opioids are needed in breastfeeding women, the lowest effective dose for the shortest duration of time should be used to limit adverse events in the mother and breastfeeding infant. In general, a single occasional dose of an opioid analgesic may be compatible with breastfeeding (WHO 2002). Breastfeeding women using opioids for postpartum pain or for the treatment of chronic maternal pain should monitor their infants for drowsiness, sedation, feeding difficulties, or limpness (ACOG 209 2019). Withdrawal symptoms may occur when maternal use is discontinued or breastfeeding is stopped.

Monitoring Parameters

Pain relief, respiratory and mental status/alertness (especially in patients on concomitant CNS depressants, including benzodiazepines), blood pressure, heart rate; blood glucose if hypoglycemia is suspected; signs/symptoms of hyponatremia (eg, confusion, disorientation) especially during initiation of therapy; bowel function; signs/symptoms of tolerance, addiction, abuse, misuse, or suicidal ideation; signs or symptoms of hypogonadism or hypoadrenalism (Brennan 2013); signs and symptoms of serotonin syndrome such as mental status changes (eg, agitation, hallucinations, coma), autonomic instability (eg, tachycardia, labile BP, hyperthermia), neuromuscular changes (eg, hyperreflexia, incoordination), and/or GI symptoms (eg, nausea, vomiting, diarrhea); signs and symptoms of neonatal withdrawal syndrome in infants born to mothers using opioids during pregnancy (eg. irritability, hyperactivity and abnormal sleep pattern, high pitched cry, tremor, vomiting, diarrhea and failure to gain weight); during discontinuation of therapy monitor pain control, withdrawal symptoms, mood changes, suicidal ideation, and for use of other substances.

Alternate recommendations: Chronic pain (long-term therapy outside of end-of-life or palliative care, active cancer treatment, sickle cell disease, or medication-based opioid use disorder treatment): Evaluate benefits/risks of opioid therapy within 1 to 4 weeks of treatment initiation and with dose increases. Re-evaluate benefits/risks every 3 months during therapy or more frequently in patients at increased risk of overdose or opioid use disorder. Urine drug testing is recommended prior to initiation and re-checking should be considered at least yearly (includes controlled prescription medications and illicit drugs of abuse). State prescription drug monitoring program (PDMP) data should be reviewed by clinicians prior to initiation and periodically during therapy (frequency ranging from every prescription to every 3 months) (CDC [Dowell 2016]).

Reference Range

100 to 300 ng/mL; however, serum level monitoring is not required

Mechanism of Action

Tramadol and its active metabolite (M1) binds to μ-opiate receptors in the CNS causing inhibition of ascending pain pathways, altering the perception of and response to pain; also inhibits the reuptake of norepinephrine and serotonin, which are neurotransmitters involved in the descending inhibitory pain pathway responsible for pain relief (Grond 2004)

Pharmacokinetics

Onset of action: Immediate release: Within 1 hour; Peak effect: 2 to 3 hours

Distribution: Vd: IV: 2.6 L/kg (males); 2.9 L/kg (females)

Protein binding, plasma: ~20%

Metabolism: Extensively hepatic via demethylation (mediated by CYP3A4 and CYP2D6), glucuronidation, and sulfation; has pharmacologically active metabolite formed by CYP2D6 (M1; O-desmethyl tramadol)

Bioavailability:

Immediate release: ~75%

Extended release: ~85% to 95% (as compared to immediate release)

Half-life elimination:

Immediate release: 6.3 ± 1.4 hours; active metabolite (M1): 7.4 ± 1.4 hours; prolonged in elderly

Extended-release:

Capsules: ~10 hours; active metabolite (M1): ~11 hours

Tablets: ~7.9 hours; active metabolite (M1): 8.8 hours.

Time to peak, plasma:

Immediate release: ~2 hours; active metabolite (M1): 3 hours

Extended release: ~4 to 12 hours; active metabolite (M1): ~5 to 15 hours

Excretion: Urine (~30% as unchanged drug; 60% as metabolites)

Pharmacokinetics: Additional Considerations

Altered kidney function: Decreased rate and extent of excretion.

Hepatic function impairment:

Immediate release: Metabolism is reduced in advanced cirrhosis, resulting in increased AUC and increased elimination half-life (13 hours [tramadol], 19 hours [M1]).

Extended release: Exposure is decreased ~50% with increased severity of hepatic impairment.

Older adult: Maximum serum concentration is increased and elimination half-life prolonged.

Sex:

Immediate release: Women had a 12% higher peak tramadol concentration and a 35% higher area under the curve (AUC) compared to men.

Extended release: AUC were somewhat higher in females than in males.

Note: Concentrations of tramadol were ~20% higher in “poor metabolizers” versus “extensive metabolizers,” while M1 concentrations were 40% lower.

Pricing: US

Capsule ER 24 Hour Therapy Pack (ConZip Oral)

100 mg (per each): $14.77

200 mg (per each): $19.35

300 mg (per each): $26.77

Capsule ER 24 Hour Therapy Pack (traMADol HCl ER Oral)

100 mg (per each): $9.18

200 mg (per each): $12.04

300 mg (per each): $16.65

Solution (Qdolo Oral)

5 mg/mL (per mL): $1.48

Solution (traMADol HCl Oral)

5 mg/mL (per mL): $2.38

Tablet, 24-hour (traMADol HCl ER (Biphasic) Oral)

100 mg (per each): $4.71

200 mg (per each): $7.78

300 mg (per each): $10.86

Tablet, 24-hour (traMADol HCl ER Oral)

100 mg (per each): $3.64

200 mg (per each): $6.02

300 mg (per each): $10.14

Tablets (traMADol HCl Oral)

50 mg (per each): $0.17 - $1.80

100 mg (per each): $1.93

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
  • Adamon (CR, DO, GT, HN, NI, PA, PL, PY, SV);
  • Adolonta (ES);
  • Amanda (TH);
  • Analab (LK, MY, TH);
  • Betram (PH);
  • Biodalgic (FR);
  • Bongesic (CR, DO, GT, HN, NI, PA, SV);
  • Calmador (AR);
  • Calmol (UY);
  • Contramal (BE, EG, FR, HU, IN, IT, TR);
  • Dolonil (BD);
  • Dolotral (PH);
  • Dolpar (ES);
  • Dolzam (BE, LU);
  • Domadol (ET, ZW);
  • Durodor Retard (MX);
  • Durotram (AU, NZ);
  • E-Dol (ZW);
  • Kontram XL SR (KR);
  • Koridol (HK);
  • Lafedol (AR);
  • Lodam (AU);
  • Lucidol (BD);
  • Lumidol (HR);
  • Mabron (AE, BH, CY, ET, IQ, IR, JO, KW, LB, LV, LY, MY, OM, SA, SG, SY, TH, YE);
  • Mabron SR (LV);
  • Mandolgin (DK);
  • Manol (CL);
  • Monoalgic (FR);
  • Newtram (KR);
  • Noax (SI);
  • Noax Uno (PL);
  • Nobligan (NO, SE);
  • Nomal (QA);
  • Onetram (JP);
  • Orasic (ID);
  • Painlax (TW);
  • Paxilfar (PT);
  • Pengesic (HK, SG);
  • Pengesic SR 100 (ET);
  • Potendol SR (KR);
  • Predxal (VN);
  • Radol (ID);
  • Ramado Retard (KR);
  • Ramgic (TW);
  • Rivadol (EG);
  • Romadol (BD);
  • Sefmal (HK, SG, VN);
  • Seminac (ID);
  • Seminac Continus (ID);
  • Sensitram (BR);
  • Syndol (LK);
  • Tadol (SI);
  • Takadol (FR);
  • Tamolan (TH);
  • Theradol (NL);
  • Topalgic (FR);
  • Trabar (CH);
  • Trabilin (BB, BM, BS, BZ, CR, DO, GT, GY, HN, JM, NI, PA, SR, SV, TT);
  • Tradmin (ZW);
  • Tradol (ET, ZW);
  • Tradolan (AE, BH, CY, IQ, IR, JO, KW, LB, LY, OM, SA, SE, SY, YE);
  • Tradonal (BE, PH);
  • Tradorec (GB);
  • Tradorec XL (IE);
  • Tralgit SR (SK);
  • Trama Inj (IL);
  • Tramacot (AR);
  • Tramada (MY);
  • Tramadex (IL);
  • Tramadol Slovakofarma (HU);
  • Tramadolor (LT);
  • Tramagetic (NO);
  • Tramagit (DE);
  • Tramahexal (ZA);
  • Tramake (IE);
  • Tramal (AE, AT, AU, BF, BH, BJ, CH, CI, CL, CO, CR, CU, CY, CZ, DO, EC, EE, EG, ET, FI, GH, GM, GN, GR, GT, HN, HR, IL, IQ, IR, JO, JP, KE, KW, LB, LR, LU, LY, MA, ML, MR, MT, MU, MW, NE, NG, NI, NL, NZ, OM, PA, PE, PH, PK, PL, PT, QA, RU, SA, SC, SD, SK, SL, SN, SV, SY, TH, TN, TZ, UG, VE, YE, ZA, ZM);
  • Tramal Long (EC);
  • Tramal Retard (AE, BH, KW, LB, QA, SA);
  • Tramalgin (BG);
  • Tramazac (BF, BJ, CI, ET, GH, GM, GN, IN, KE, LR, MA, ML, MR, MU, MW, NE, NG, SC, SD, SL, SN, TH, TN, TZ, UG, ZM);
  • Tramcontin (CN);
  • Tramed (TW);
  • Tramedo (AU);
  • Tramica (ZW);
  • Tramol (LK);
  • Tramundin (DE);
  • Tramundin Retard (AE, KW);
  • Tramundin retard (BH);
  • Trasic (LK);
  • Trasik (ID);
  • TRD-Contin (IN);
  • Trexol (MX);
  • Tridol (KR);
  • Trol (BD);
  • Ultradol (BD);
  • Unitral (PH);
  • Urgendol (IN);
  • Vesnon-V (TH);
  • Zamadol (BR, GB);
  • Zamudol (FR);
  • Zodol (CL, PY);
  • Zudol (LK);
  • Zydol (AU, GB, IE);
  • Zydol XL (GB);
  • Zytram (ES);
  • Zytram XL SR (KR)


For country code abbreviations (show table)
  1. 2019 American Geriatrics Society Beers Criteria Update Expert Panel. American Geriatrics Society 2019 updated AGS Beers Criteria for Potentially Inappropriate Medication Use in Older Adults. J Am Geriatr Soc. 2019;67(4):674-694. doi:10.1111/jgs.15767 [PubMed 30693946]
  2. Agrawal A, Diwan SK, Mahajan R. Severe delirium following single dose of tramadol. Indian J Med Sci. 2009;63(2):80-81. doi:10.4103/0019-5359.49246 [PubMed 19359773]
  3. Alghobary M, El-Bayoumy Y, Mostafa Y, Mahmoud el-HM, Amr M. Evaluation of tramadol on demand vs daily paroxetine as a long-term treatment of lifelong premature ejaculation. J Sex Med. 2010;7(8):2860-2867. doi:10.1111/j.1743-6109.2010.01789.x [PubMed 20367773]
  4. Althof SE, McMahon CG, Waldinger MD, et al. An update of the International Society of Sexual Medicine's guidelines for the diagnosis and treatment of premature ejaculation (PE). J Sex Med. 2014;11(6):1392-1422. [PubMed 24848686]
  5. American College of Obstetricians and Gynecologists (ACOG). ACOG practice bulletin no. 209: obstetric analgesia and anesthesia. Obstet Gynecol. 2019;133(3):e208-e225. [PubMed 30801474]
  6. American Dental Association statement on the use of opioids in the treatment of dental pain. 2016. https://www.ada.org/en/about-the-ada/ada-positions-policies-and-statements/substance-use-disorders
  7. American Dental Association ADA adopts interim opioids policy 2018. https://www.ada.org/en/publications/ada-news/2018-archive/march/ada-adopts-interim-opioids-policy
  8. American Pain Society. Principles of Analgesic Use. 7th ed. American Pain Society; 2016.
  9. Aminoshariae A, Kulild JC, Donaldson M, Hersh EV. Evidence-based recommendations for analgesic efficacy to treat pain of endodontic origin: A systematic review of randomized controlled trials. J Am Dent Assoc. 2016;147(10):826-839. [PubMed 27475974]
  10. Anderson PO, Sauberan JB. Modeling drug passage into human milk. Clin Pharmacol Ther. 2016;100(1):42-52. [PubMed 27060684]
  11. Bachs LC, Engeland A, Mørland JG, Skurtveit S. The risk of motor vehicle accidents involving drivers with prescriptions for codeine or tramadol. Clin Pharmacol Ther. 2009;85(6):596-599. [PubMed 19279562]
  12. Bandieri E, Romero M, Ripamonti CI, et al. Randomized trial of low-dose morphine versus weak opioids in moderate cancer pain. J Clin Oncol. 2016;34(5):436-442. doi:10.1200/JCO.2015.61.0733 [PubMed 26644526]
  13. Based on expert opinion.
  14. Bassiony MM, Youssef UM, Hassan MS, et al. Cognitive impairment and tramadol dependence. J Clin Psychopharmacol. 2017;37(1):61-66. doi:10.1097/JCP.0000000000000617 [PubMed 27918316]
  15. Beakley BD, Kaye AM, Kaye AD. Tramadol, pharmacology, side effects, and serotonin syndrome: A review. Pain Physician. 2015;18(4):395-400. [PubMed 26218943]
  16. Berna C, Kulich RJ, Rathmell JP. Tapering long-term opioid therapy in chronic noncancer pain: evidence and recommendations for everyday practice. Mayo Clin Proc. 2015;90(6):828-842. doi:10.1016/j.mayocp.2015.04.003 [PubMed 26046416]
  17. Bosilkovska M, Walder B, Besson M, Daali Y, Desmeules J. Analgesics in patients with hepatic impairment: pharmacology and clinical implications. Drugs. 2012;72(12):1645-1669. doi:10.2165/11635500-000000000-00000 [PubMed 22867045]
  18. Brennan MJ. The effect of opioid therapy on endocrine function. Am J Med. 2013;126(3)(suppl 1):S12-S18. doi:10.1016/j.amjmed.2012.12.001 [PubMed 23414717]
  19. Burma NE, Kwok CH, Trang T. Therapies and mechanisms of opioid withdrawal. Pain Manag. 2017;7(6):455-459. doi:10.2217/pmt-2017-0028 [PubMed 29125396]
  20. Busse J; National Pain Center. The 2017 Canadian guideline for opioids for chronic non-cancer pain. http://nationalpaincentre.mcmaster.ca/guidelines.html. Published 2017. Accessed September 13, 2018 [PubMed 28483845]
  21. Centers for Disease Control and Prevention (CDC). Common elements in guidelines for prescribing opioids for chronic pain. https://www.cdc.gov/drugoverdose/pdf/common_elements_in_guidelines_for_prescribing_opioids-a.pdf. Published 2015. Accessed September 13, 2018.
  22. Chandok N, Watt KD. Pain management in the cirrhotic patient: the clinical challenge. Mayo Clin Proc. 2010;85(5):451-458. doi:10.4065/mcp.2009.0534 [PubMed 20357277]
  23. Chou R, Fanciullo GJ, Fine PG, et al. Clinical Guidelines for the Use of Chronic Opioid Therapy in Chronic Noncancer Pain. J Pain. 2009;10(2):113-130. [PubMed 19187889]
  24. ConZip (tramadol) extended-release capsules [prescribing information]. Alpharetta, GA: Vertical Pharmaceuticals LLC; January 2022.
  25. Crews KR, Monte AA, Huddart R, et al. Clinical Pharmacogenetics Implementation Consortium guideline for CYP2D6, OPRM1, and COMT genotypes and select opioid therapy. Clin Pharmacol Ther. 2021;110(4):888-896. doi:10.1002/cpt.2149 [PubMed 33387367]
  26. Dahan A, van der Schrier R, Smith T, Aarts L, van Velzen M, Niesters M. Averting opioid-induced respiratory depression without affecting analgesia. Anesthesiology. 2018;128(5):1027-1037. doi:10.1097/ALN.0000000000002184 [PubMed 29553984]
  27. Davison SN, Koncicki H, Brennan F. Pain in chronic kidney disease: a scoping review. Semin Dial. 2014;27(2):188-204. doi:10.1111/sdi.12196 [PubMed 24517512]
  28. Debono M, Chan S, Rolfe C, Jones TH. Tramadol-induced adrenal insufficiency. Eur J Clin Pharmacol. 2011;67(8):865-867. doi:10.1007/s00228-011-0992-9 [PubMed 21243342]
  29. Doroschak AM, Bowles WR, Hargreaves KM. Evaluation of the combination of flurbiprofen and tramadol for management of endodontic pain. J Endod. 1999;25(10):660-663. [PubMed 10687523]
  30. Dow K, Ordean A, Murphy-Oikonen J, et al. Neonatal Abstinence Syndrome Clinical Practice Guidelines for Ontario. J Popul Ther Clin Pharmacol. 2012;19(3):e488-e506. [PubMed 23241498]
  31. Dowell D. CDC issues key clarification on guideline for prescribing opioids for chronic pain. https://www.asco.org/advocacy-policy/asco-in-action/cdc-issues-key-clarification-guideline-prescribing-opioids-chronic. Published February 28, 2019. Accessed June 28, 2019.
  32. Dowell D, Haegerich TM, Chou R. CDC guideline for prescribing opioids for chronic pain—United States, 2016. MMWR Recomm Rep. 2016;65(1):1-49. doi:10.15585/mmwr.rr6501e1 [PubMed 26987082]
  33. Dunn KE, Bergeria CL, Huhn AS, Strain EC. A systematic review of laboratory evidence for the abuse potential of tramadol in humans. Front Psychiatry. 2019;10:704. doi:10.3389/fpsyt.2019.00704 [PubMed 31616329]
  34. Dunn K, Bergeria C, Huhn AS, Strain EC. Differences in patient-reported and observer-rated opioid withdrawal symptom etiology, time course, and relationship to clinical outcome. Drug Alcohol Depend. 2020;215:108212. doi:10.1016/j.drugalcdep.2020.108212 [PubMed 32781310]
  35. Durela (tramadol) [product monograph]. Mississauga, Ontario, Canada: Cipher Pharmaceuticals Inc; March 2022.
  36. Dwyer JP, Jayasekera C, Nicoll A. Analgesia for the cirrhotic patient: a literature review and recommendations. J Gastroenterol Hepatol. 2014;29(7):1356-1360. doi:10.1111/jgh.12560 [PubMed 24548074]
  37. Eassa BI, El-Shazly MA. Safety and efficacy of tramadol hydrochloride on treatment of premature ejaculation. Asian J Androl. 2013;15(1):138-142. doi:10.1038/aja.2012.96 [PubMed 23103596]
  38. El-Hadidy MA, Helaly AM. Medical and psychiatric effects of long-term dependence on high dose of tramadol. Subst Use Misuse. 2015;50(5):582-589. doi:10.3109/10826084.2014.991406 [PubMed 25544109]
  39. Finkel JC, Rose JB, Schmitz ML, et al. An Evaluation of the Efficacy and Tolerability of Oral Tramadol Hydrochloride Tablets for the Treatment of Postsurgical Pain in Children. Anesth Analg. 2002;94(6):1469-1473. [PubMed 12032009]
  40. Food and Drug Administration (FDA). FDA Drug Safety Communication: FDA restricts use of prescription codeine pain and cough medicines and tramadol pain medicines in children; recommends against use in breastfeeding women. 2017. https://www.fda.gov/Drugs/DrugSafety/ucm549679.htm
  41. Food and Drug Administration (FDA). Tramadol: drug safety communication - FDA evaluating risks of using in children aged 17 and younger. 2015. Available at https://www.fda.gov/safety/medwatch/safetyinformation/safetyalertsforhumanmedicalproducts/ucm463499.htm
  42. Fournier JP, Azoulay L, Yin H, Montastruc JL, Suissa S. Tramadol use and the risk of hospitalization for hypoglycemia in patients with noncancer pain. JAMA Intern Med. 2015;175(2):186-193. doi:10.1001/jamainternmed.2014.6512 [PubMed 25485799]
  43. Fricke JR Jr, Hewitt DJ, Jordan DM, Fisher A, Rosenthal NR. A double-blind placebo-controlled comparison of tramadol/acetaminophen and tramadol in patients with postoperative dental pain. Pain. 2004;109(3):250-257. [PubMed 15157685]
  44. Gameel TA, Tawfik AM, Abou-Farha MO, Bastawisy MG, El-Bendary MA, El-Gamasy Ael-N. On-demand use of tramadol, sildenafil, paroxetine and local anaesthetics for the management of premature ejaculation: A randomised placebo-controlled clinical trial. Arab J Urol. 2013;11(4):392-397. doi:10.1016/j.aju.2013.05.003 [PubMed 26558110]
  45. Gibson TP. Pharmacokinetics, efficacy, and safety of analgesia with a focus on tramadol HCl. Am J Med. 1996;101(1A):47S-53S. doi:10.1016/s0002-9343(96)00138-6 [PubMed 8764760]
  46. Grond S, Sablotzki A. Clinical pharmacology of tramadol. Clin Pharmacokinet. 2004;43(13):879-923. [PubMed 15509185]
  47. Hansen CA, Ernst MT, Stougaard M, Abrahamsen B. Tramadol prescribed use in general and chronic noncancer pain: a nationwide register-based cohort study of all patients above 16 years. Scand J Pain. 2019;20(1):109-124. doi:10.1515/sjpain-2019-0114 [PubMed 31527300]
  48. Hassamal S, Miotto K, Dale W, Danovitch I. Tramadol: understanding the risk of serotonin syndrome and seizures. Am J Med. 2018;131(11):1382.e1-1382.e6. doi:10.1016/j.amjmed.2018.04.025 [PubMed 29752906]
  49. Hassanian-Moghaddam H, Farajidana H, Sarjami S, Owliaey H. Tramadol-induced apnea. Am J Emerg Med. 2013;31(1):26-31. doi:10.1016/j.ajem.2012.05.013 [PubMed 22809771]
  50. Hill MV, Stucke RS, McMahon ML, Beeman JL, Barth RJ Jr. An educational intervention decreases opioid prescribing after general surgical operations. Ann Surg. 2018;267(3):468-472. doi:10.1097/SLA.0000000000002198 [PubMed 28267689]
  51. Hudak ML, Tan RC; Committee On Drugs, et al. Neonatal Drug Withdrawal. Pediatrics. 2012;129(2):e540-e560. [PubMed 22291123]
  52. Hull MJ, Griggs D, Knoepp SM, et al. Postmortem Urine Immunoassay Showing False-Positive Phencyclidine Reactivity in a Case of Fatal Tramadol Overdose. Am J Forensic Med Pathol. 2006;27(4):359-362.
  53. Ilett KF, Paech MJ, Page-Sharp M, et al. Use of a sparse sampling study design to assess transfer of tramadol and its O-desmethyl metabolite into transitional breast milk. Br J Clin Pharmacol. 2008;65(5):661-666. [PubMed 18294329]
  54. Isaac Z, Atlas S, Kunis L. Management of non-radicular neck pain in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed June 22, 2022.
  55. Ito S. Drug therapy for breast-feeding women. N Engl J Med. 2000;343(2):118-126. [PubMed 10891521]
  56. JAMP Tramadol (tramadol) [product monograph]. Boucherville, Quebec, Canada: JAMP Pharma Corp; March 2022.
  57. Javelot H, Michel B, Marquis A, et al. Acute withdrawal syndrome after discontinuation of a short analgesic treatment with tramadol. Therapie. 2016;71(3):347-348. doi:10.1016/j.therap.2015.11.007 [PubMed 27235663]
  58. Kaplan A, Rosenblatt R. Symptom management in patients with cirrhosis: a practical guide. Curr Treat Options Gastroenterol. 2022;20(2):144-159. doi:10.1007/s11938-022-00377-y [PubMed 35313484]
  59. Kaynar M, Kilic O, Yurdakul T. On-demand tramadol hydrochloride use in premature ejaculation treatment. Urology. 2012;79(1):145-149. doi:10.1016/j.urology.2011.09.031 [PubMed 22088571]
  60. King S, Forbes K, Hanks GW, Ferro CJ, Chambers EJ. A systematic review of the use of opioid medication for those with moderate to severe cancer pain and renal impairment: a European Palliative Care Research Collaborative opioid guidelines project. Palliat Med. 2011;25(5):525-552. doi:10.1177/0269216311406313 [PubMed 21708859]
  61. Koncicki HM, Brennan F, Vinen K, Davison SN. An approach to pain management in end stage renal disease: considerations for general management and intradialytic symptoms. Semin Dial. 2015;28(4):384-391. doi:10.1111/sdi.12372 [PubMed 25864854]
  62. Koncicki HM, Unruh M, Schell JO. Pain management in CKD: a guide for nephrology providers. Am J Kidney Dis. 2017;69(3):451-460. doi:10.1053/j.ajkd.2016.08.039 [PubMed 27881247]
  63. Krčevski Škvarč N, Morlion B, Vowles KE, et al. European clinical practice recommendations on opioids for chronic noncancer pain - part 2: special situations. Eur J Pain. 2021;25(5):969-985. doi:10.1002/ejp.1744 [PubMed 33655678]
  64. Künig G, Dätwyler S, Eschen A, Schreiter Gasser U. Unrecognised long-lasting tramadol-induced delirium in two elderly patients. A case report. Pharmacopsychiatry. 2006;39(5):194-199. doi:10.1055/s-2006-948331 [PubMed 16944413]
  65. Kurella M, Bennett WM, Chertow GM. Analgesia in patients with ESRD: a review of available evidence. Am J Kidney Dis. 2003;42(2):217-228. doi:10.1016/s0272-6386(03)00645-0 [PubMed 12900801]
  66. Langley PC, Patkar AD, Boswell KA, Benson CJ, Schein JR. Adverse event profile of tramadol in recent clinical studies of chronic osteoarthritis pain. Curr Med Res Opin. 2010;26(1):239-251. doi:10.1185/03007990903426787 [PubMed 19929615]
  67. Lanier RK, Lofwall MR, Mintzer MZ, Bigelow GE, Strain EC. Physical dependence potential of daily tramadol dosing in humans. Psychopharmacology (Berl). 2010;211(4):457-466. doi:10.1007/s00213-010-1919-3 [PubMed 20589494]
  68. Lewis JH, Stine JG. Review article: prescribing medications in patients with cirrhosis - a practical guide. Aliment Pharmacol Ther. 2013;37(12):1132-1156. doi:10.1111/apt.12324 [PubMed 23638982]
  69. Makris A, Matala ME, Tsirigotis A, Karmaniolou I. Apnea and mydriasis after postoperative tramadol administration: an unusual complication and possible underlying mechanisms. Anaesthesia. 2012;67(1):76-77. doi:10.1111/j.1365-2044.2011.06969.x [PubMed 22150490]
  70. 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. [PubMed 29595994]
  71. Martyn-St James M, Cooper K, Kaltenthaler E, et al. Tramadol for premature ejaculation: a systematic review and meta-analysis. BMC Urol. 2015;15:6. [PubMed 25636495]
  72. Mehrvarzfar P, Abbott PV, Saghiri MA, Delvarani A, Asgar K, Lotfi M, Karamifar K, Kharazifard MJ, Khabazi H. Effects of three oral analgesics on postoperative pain following root canal preparation: a controlled clinical trial. Int Endod J. 2012;45(1):76-82. [PubMed 21902704]
  73. Memarian A, Farhidnia N, Fallahi F. Generalized tonic colonic seizure followed by loss of consciousness early after using low dose of tramadol: A case report. Anesth Pain Med. 2018;8(3):e64707. doi:10.5812/aapm.64707 [PubMed 30214882]
  74. Meyers RS, Thackray J, Matson KL, et al. Key Potentially Inappropriate Drugs in Pediatrics: The KIDs List. J Pediatr Pharmacol Ther. 2020;25(3):175-191. [PubMed 32265601]
  75. Miotto K, Cho AK, Khalil MA, Blanco K, Sasaki JD, Rawson R. Trends in tramadol: pharmacology, metabolism, and misuse. Anesth Analg. 2017;124(1):44-51. doi:10.1213/ANE.0000000000001683 [PubMed 27861439]
  76. Mishra H, Khan FA. A double-blind, placebo-controlled randomized comparison of pre and postoperative administration of ketorolac and tramadol for dental extraction pain. J Anaesthesiol Clin Pharmacol. 2012;28(2):221-225. [PubMed 22557747]
  77. Mockenhaupt M, Viboud C, Dunant A, et al. Stevens-Johnson syndrome and toxic epidermal necrolysis: assessment of medication risks with emphasis on recently marketed drugs. The EuroSCAR-study. J Invest Dermatol. 2008;128(1):35-44. doi:10.1038/sj.jid.5701033 [PubMed 17805350]
  78. Moore PA, Ziegler KM, Lipman RD, Aminoshariae A, Carrasco-Labra A, Mariotti A. Benefits and harms associated with analgesic medications used in the management of acute dental pain: an overview of systematic reviews. J Am Dent Assoc. 2018;149(4):256-265.e3. [PubMed 29599019]
  79. Mori F, Barni S, Manfredi M, et al. Anaphylaxis to intravenous tramadol in a child. Pharmacology. 2015;96(5-6):256-258. doi:10.1159/000441005 [PubMed 26550831]
  80. Nadpara PA, Joyce AR, Murrelle EL, et al. Risk factors for serious prescription opioid-induced respiratory depression or overdose: comparison of commercially insured and veterans health affairs populations. Pain Med. 2018;19(1):79-96. doi:10.1093/pm/pnx038 [PubMed 28419384]
  81. Nakhaee S, Amirabadizadeh A, Brent J, et al. Tramadol and the occurrence of seizures: a systematic review and meta-analysis. Crit Rev Toxicol. 2019;49(8):710-723. doi:10.1080/10408444.2019.1694861 [PubMed 31914355]
  82. Nelson AD, Camilleri M. Opioid-induced constipation: advances and clinical guidance. Ther Adv Chronic Dis. 2016;7(2):121-134. doi:10.1177/2040622315627801 [PubMed 26977281]
  83. Nelson EM, Philbrick AM. Avoiding serotonin syndrome: the nature of the interaction between tramadol and selective serotonin reuptake inhibitors. Ann Pharmacother. 2012;46(12):1712-1716. doi:10.1345/aph.1Q748 [PubMed 23212934]
  84. Odonkor CA, Chhatre A. What's tramadol got to do with it? A case report of rebound hypoglycemia, a reappraisal and review of potential mechanisms. Pain Physician. 2016;19(8):E1215-E1220. [PubMed 27906953]
  85. Orliaguet G, Hamza J, Couloigner V, et al. A case of respiratory depression in a child with ultrarapid CYP2D6 metabolism after tramadol. Pediatrics. 2015;135(3):e753-755. [PubMed 25647677 ]
  86. Park YM, Park HK, Kim L, Lee HJ, Kang SG. Acute-withdrawal restless legs syndrome following abrupt cessation of short-term tramadol. Psychiatry Investig. 2014;11(2):204-206. doi:10.4306/pi.2014.11.2.204 [PubMed 24843378]
  87. Payne KA, Roelofse JA, Shipton EA. Pharmacokinetics of Oral Tramadol Drops for Postoperative Pain Relief in Children Aged 4 to 7 Years − A Pilot Study. Anesth Prog. 2002;49(4):109-112. [PubMed 12779111]
  88. Pham PC, Khaing K, Sievers TM, et al. 2017 update on pain management in patients with chronic kidney disease. Clin Kidney J. 2017;10(5):688-697. doi:10.1093/ckj/sfx080 [PubMed 28979781]
  89. Pino CA, Covington M, Wakeman SE. Prescription of opioids for acute pain in opioid naïve patients. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed January 25, 2022.
  90. Prakash J, Saini R. Tramadol dependence: a case report. Med J Armed Forces India. 2010;66(1):93-94. doi:10.1016/S0377-1237(10)80114-5 [PubMed 27365718]
  91. Qdolo (tramadol) [prescribing information]. Athens, GA: Athena Bioscience LLC; September 2021.
  92. Ralivia (tramadol) [product monograph]. Laval, Quebec, Canada: Bausch Health, Canada Inc; March 2022.
  93. Reece-Stremtan S, Campos M, Kokajko L; Academy of Breastfeeding Medicine. ABM clinical protocol #15: analgesia and anesthesia for the breastfeeding other, revised 2017. Breastfeed Med. 2017;12(9):500-506. [PubMed 29624435]
  94. Refer to manufacturer labeling.
  95. Rehni AK, Singh I, Kumar M. Tramadol-induced seizurogenic effect: a possible role of opioid-dependent gamma-aminobutyric acid inhibitory pathway. Basic Clin Pharmacol Toxicol. 2008;103(3):262-266. doi:10.1111/j.1742-7843.2008.00276.x [PubMed 18684224]
  96. Rehni AK, Singh TG, Singh N, Arora S. Tramadol-induced seizurogenic effect: a possible role of opioid-dependent histamine H1 receptor activation-linked mechanism. Naunyn Schmiedebergs Arch Pharmacol. 2010;381(1):11-19. doi:10.1007/s00210-009-0476-y [PubMed 20012267]
  97. Rogal SS, Hansen L, Patel A, et al. AASLD practice guidance: palliative care and symptom-based management in decompensated cirrhosis. Hepatology. 2022;76(3):819-853. doi:10.1002/hep.32378 [PubMed 35103995]
  98. Rose JB, Finkel JC, Arquedas-Mohs A, et al. Oral Tramadol for the Treatment of Pain of 7-30 Days' Duration in Children. Anesth Analg. 2003;96(1):78-81. [PubMed 12505927]
  99. Rosenquist R. Use of opioids in the management of chronic non-cancer pain. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed May 31, 2022.
  100. Rougemont-Bücking A, Gamma F, Panksepp J. Use of tramadol in psychiatric care: a comprehensive review and report of two cases. Swiss Med Wkly. 2017;147:w14428. doi:10.4414/smw.2017.14428 [PubMed 28488261]
  101. Roussin A, Doazan-d'Ouince O, Géniaux H, Halberer C; French Network of Centre for Evaluation and Information on Pharmacodependence (Addictovigilance Centres). Evaluation of abuse and dependence in addiction monitoring systems: tramadol as an example. Article in English and French. Therapie. 2015;70(2):203-221. doi:10.2515/therapie/2015014 [PubMed 25858577]
  102. Rudisill TM, Zhu M, Davidov D, et al. Medication use and the risk of motor vehicle collision in West Virginia drivers 65 years of age and older: a case-crossover study. BMC Res Notes. 2016a;9:166. [PubMed 26979111]
  103. Rudisill TM, Zhu M, Kelley GA, Pilkerton C, Rudisill BR. Medication use and the risk of motor vehicle collisions among licensed drivers: A systematic review. Accid Anal Prev. 2016b;96:255-270. [PubMed 27569655]
  104. Sachs HC; Committee On Drugs. The transfer of drugs and therapeutics into human breast milk: an update on selected topics. Pediatrics. 2013;132(3):e796-e809. [PubMed 23979084]
  105. Safarinejad MR, Hosseini SY. Safety and efficacy of tramadol in the treatment of premature ejaculation: a double-blind, placebo-controlled, fixed-dose, randomized study. J Clin Psychopharmacol. 2006;26(1):27-31. [PubMed 16415702]
  106. Salem EA, Wilson SK, Bissada NK, Delk JR, Hellstrom WJ, Cleves MA. Tramadol HCl has promise in on-demand use to treat premature ejaculation. J Sex Med. 2008;5(1):188-193. [PubMed 17362279]
  107. Salman S, Sy SK, Ilett KF, Page-Sharp M, Paech MJ. Population pharmacokinetic modeling of tramadol and its O-desmethyl metabolite in plasma and breast milk. Eur J Clin Pharmacol. 2011;67(9):899-908. [PubMed 21394525]
  108. Sánchez-González MJ, Barbarroja-Escudero J, Antolín-Amérigo D, et al. Erythema multiforme induced by tramadol: an allergy assessment. J Investig Allergol Clin Immunol. 2020;30(4):290-291. doi:10.18176/jiaci.0495 [PubMed 32044727]
  109. Sarkar S, Nebhinani N, Singh SM, Mattoo SK, Basu D. Tramadol dependence: a case series from India. Indian J Psychol Med. 2012;34(3):283-285. doi:10.4103/0253-7176.106038 [PubMed 23440178]
  110. Schiemsky T, Vundelinckx G, Croes K, et al. An unconscious man with profound drug-induced hypoglycaemia. Biochem Med (Zagreb). 2020;30(1):010802. doi:10.11613/BM.2020.010802 [PubMed 31839727]
  111. Senay EC, Adams EH, Geller A, et al. Physical dependence on Ultram (tramadol hydrochloride): both opioid-like and atypical withdrawal symptoms occur. Drug Alcohol Depend. 2003;69(3):233-241. doi:10.1016/s0376-8716(02)00321-6 [PubMed 12633909]
  112. Sevarino K. Medically supervised opioid withdrawal during treatment for addiction. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed September 13, 2018.
  113. Shakoor M, Ayub S, Ahad A, Ayub Z. Transient serotonin syndrome caused by concurrent use of tramadol and selective serotonin reuptake inhibitor. Am J Case Rep. 2014;15:562-564. doi:10.12659/AJCR.892264 [PubMed 25540831]
  114. Shindel AW, Althof SE, Carrier S, et al. Disorders of ejaculation: an AUA/SMSNA guideline. J Urol. 2022;207(3):504-512. doi:10.1097/JU.0000000000002392 [PubMed 34961344]
  115. Smith HS. The metabolism of opioid agents and the clinical impact of their active metabolites. Clin J Pain. 2011;27(9):824-838. doi:10.1097/AJP.0b013e31821d8ac1 [PubMed 21677572]
  116. Soleimanpour H, Safari S, Shahsavari Nia K, Sanaie S, Alavian SM. Opioid drugs in patients with liver disease: a systematic review. Hepat Mon. 2016;16(4):e32636. doi:10.5812/hepatmon.32636 [PubMed 27257423]
  117. Stamer UM, Stüber F, Muders T, Musshoff F. Respiratory depression with tramadol in a patient with renal impairment and CYP2D6 gene duplication. Anesth Analg. 2008;107(3):926-939. doi:10.1213/ane.0b013e31817b796e [PubMed 18713907]
  118. Tashakori A, Afshari R. Tramadol overdose as a cause of serotonin syndrome: a case series. Clin Toxicol (Phila). 2010;48(4):337-341. doi:10.3109/15563651003709427 [PubMed 20367390]
  119. Tramadol Hydrochloride ER tablets [prescribing information]. Morgantown, WV: Mylan Pharmaceuticals Inc; June 2021.
  120. Tridural (tramadol) [product monograph]. Saint-Laurent, Quebec, Canada: Paladin Labs Inc; March 2022.
  121. Ultram (tramadol) [prescribing information]. Titusville, NJ: Janssen Pharmaceuticals Inc; September 2021.
  122. Ultram (tramadol) [product monograph]. Toronto, Ontario, Canada: Janssen Inc; March 2022.
  123. Ultram ER (tramadol) [prescribing information]. Titusville, NJ: Janssen Pharmaceuticals Inc; August 2017.
  124. Wei J, Lane NE, Bolster MB, et al. Association of tramadol use with risk of hip fracture. J Bone Miner Res. Published online February 5, 2020. [PubMed 32020683]
  125. Wiffen PJ, Derry S, Moore RA. Tramadol with or without paracetamol (acetaminophen) for cancer pain. Cochrane Database Syst Rev. 2017;5:CD012508. doi:10.1002/14651858.CD012508.pub2 [PubMed 28510996]
  126. Wilcock A, Charlesworth S, Prentice W, et al. Prescribing in chronic severe hepatic impairment. J Pain Symptom Manage. 2019;58(3):515-537. doi:10.1016/j.jpainsymman.2019.04.034 [PubMed 31077785]
  127. World Health Organization (WHO). Breastfeeding and maternal medication, recommendations for drugs in the Eleventh WHO Model List of Essential Drugs. 2002. http://www.who.int/maternal_child_adolescent/documents/55732/en/
  128. Zytram XL (tramadol) [product monograph]. Toronto, Ontario, Canada: Purdue Pharma; March 2022.
Topic 10010 Version 609.0