ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Oxycodone: Pediatric drug information

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

For abbreviations, symbols, and age group definitions used in Lexicomp (show table)
Special Alerts
FDA Requiring Updates to Opioid Prescribing Information April 2023

The FDA has issued a drug safety communication to announce safety-related updates to the prescribing information for immediate-release (IR) and extended-release (ER)/long-acting (LA) opioid analgesics, including updates to Boxed Warnings, Indications and Usage, Dosage and Administration, Warnings and Precautions, and the Medication Guide. These safety labeling changes are intended to provide clarity on appropriate patient populations for opioid treatment, appropriate dosage and administration, and updated information on the risks associated with opioid use. The required safety labeling changes include stating:

  • the risk of overdose increases as the dosage increases for all opioid pain medicines;

  • IR opioids should not be used for an extended period of time unless a patient's pain remains severe enough to require them and alternative treatment options continue to be inadequate;

  • many acute pain conditions treated in the outpatient setting require no more than a few days of an opioid pain medicine;

  • it is recommended to reserve ER/LA opioid pain medicines for severe and persistent pain that requires an extended treatment period with a daily opioid pain medicine and for which alternative treatment options are inadequate; and

  • a warning about opioid-induced hyperalgesia (OIH), including information on differentiating OIH symptoms from those of opioid tolerance and withdrawal.

Further information may be found at https://www.fda.gov/drugs/drug-safety-and-availability/fda-updates-prescribing-information-all-opioid-pain-medicines-provide-additional-guidance-safe-use.

ALERT: US Boxed Warning
Addiction, abuse, and misuse:

Oxycodone 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 oxycodone and monitor all patients regularly for the development of these behaviors or 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 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 health care providers. Health care providers are strongly encouraged to complete a REMS-compliant education program and 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 oxycodone. Monitor for respiratory depression, especially during initiation of oxycodone or following a dose increase. Instruct patients to swallow oxycodone tablets whole; crushing, chewing, or dissolving oxycodone ER tablets can cause rapid release and absorption of a potentially fatal dose of oxycodone.

Accidental ingestion:

Accidental ingestion of even one dose of oxycodone, especially by children, can result in a fatal overdose of oxycodone.

Neonatal opioid withdrawal:

Prolonged use of oxycodone 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.

Cytochrome P450 3A4 interaction:

The concomitant use of oxycodone with all cytochrome P450 (CYP-450) 3A4 inhibitors may result in an increase in oxycodone plasma concentrations, which could increase or prolong adverse drug effects and may cause potentially fatal respiratory depression. In addition, discontinuation of a concomitantly used CYP3A4 inducer may result in an increase in oxycodone plasma concentration. Monitor patients receiving oxycodone and any CYP3A4 inhibitor or inducer.

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 oxycodone and benzodiazepines or other CNS depressants for use in patients for whom alternative treatment options are inadequate. Limit dosages and durations to the minimum required. Follow patients for signs and symptoms of respiratory depression and sedation.

Risk of medication errors (oral solution):

Ensure accuracy when prescribing, dispensing, and administering oxycodone oral solution. Dosing errors due to confusion between mg and mL, and other oxycodone oral solutions of different concentrations can result in accidental overdose.

Brand Names: US
  • Oxaydo;
  • OxyCONTIN;
  • Roxicodone;
  • RoxyBond;
  • Xtampza ER
Brand Names: Canada
  • ACT Oxycodone CR [DSC];
  • APO-Oxycodone CR;
  • Oxy-IR;
  • OxyNEO;
  • PMS-OxyCODONE;
  • PMS-OxyCODONE CR;
  • Supeudol;
  • Supeudol 10;
  • Supeudol 20
Therapeutic Category
  • Analgesic, Opioid
Dosing: Pediatric

Dosage guidance:

Safety: Doses should be titrated to appropriate effect. Multiple concentrations of oral solution available (20 mg/mL and 1 mg/mL); the highly concentrated formulation (20 mg/mL) should only be used in opioid-tolerant patients (taking ≥30 mg/day of oxycodone or equivalent for ≥1 week). Orders for oxycodone oral solutions (20 mg/mL or 1 mg/mL) should be clearly written to include the intended dose (in mg not mL) and the intended product concentration to be dispensed to avoid potential dosing errors:

Analgesia, moderate to severe pain

Analgesia, moderate to severe pain: Limited data available:

Infants ≤6 months: Immediate release: Oral solution (1 mg/mL): Oral: Initial dose: 0.025 to 0.05 mg/kg/dose every 4 to 6 hours as needed (Ref).

Infants >6 months, Children, and Adolescents:

Patient weight <50 kg: Immediate release: Oral: Initial dose: 0.1 to 0.2 mg/kg/dose every 4 to 6 hours as needed; for severe pain some experts have recommended an initial dose of 0.2 mg/kg; usual maximum dose range: 5 to 10 mg/dose (Ref).

Patient weight ≥50 kg: Immediate release: Oral: Initial dose: 5 to 10 mg every 4 to 6 hours as needed; for severe pain an initial dose of 10 mg may be used; usual maximum dose: 20 mg/dose (Ref).

Analgesia, severe pain requiring around-the-clock long-term opioid therapy

Analgesia, severe pain requiring around-the-clock long-term opioid therapy: Note: Use only in pediatric patients ≥11 years of age who are already receiving opioid therapy for ≥5 consecutive days, tolerating a minimum daily opioid dose of ≥20 mg of oxycodone orally or its equivalent at least for the 2 days immediately prior to starting extended-release oxycodone tablets, and for which alternative treatment options are inadequate. Prior to initiation, all other around-the-clock opioid therapy must be discontinued.

Children ≥11 years and Adolescents: Extended-release tablets (eg, OxyContin): Oral:

Initial dose: Based on current opioid regimen dose; use the following conversion factor table and equation to convert the current opioid(s) daily dose to the extended-release oxycodone tablet daily dose.

Note: Substantial interpatient variability exists due to patient specific factors, relative potency of different opioids, and dosage forms; therefore, it is preferable to underestimate the initial 24-hour oral oxycodone requirements and utilize rescue medication (immediate-release opioid):

Initial dose of extended-release oxycodone tablets every 12 hours = (mg/day of current opioid regimen X conversion factor)/2

Dose calculations or adjustments for specific clinical scenarios:

• If rounding is necessary, numerical value should be rounded down to the nearest tablet strength. If calculated daily dose is <20 mg, do not start extended-release oxycodone tablet as there is no safe tablet strength available.

• If more than one opioid in the regimen, calculate the approximate extended-release oxycodone tablet dose for each opioid and sum the totals for the approximate total daily extended-release oxycodone tablet dose, then divide by 2 for the 12-hour extended-release oxycodone dose.

• If current opioid regimen includes a fixed-dose opioid/nonopioid dosage form (eg, hydrocodone/acetaminophen), only the mg of opioid should be used in the conversion calculations.

• If patient receiving concomitant CNS depressants, reduce extended-release oxycodone tablet starting dose by 1/3 to 1/2 the calculated initial dose.

• If asymmetric dosing, the higher dose should be scheduled as the morning dose, and the lower dose 12 hours later.

Note: The following conversion table should ONLY be used to convert opioid doses to extended-release oxycodone tablet (not from extended-release oxycodone tablet to other opioids; it is NOT a table of equianalgesic doses as it may overestimate initial dose).

Conversion Factor for Calculating Initial Extended-Release Oxycodone Tablet Dose in Pediatric Patients ≥11 Years

Current opioid regimen to be converted to extended-release oxycodone tablet

Conversion factor

Oral

Parenterala

Oxycodone

1

--

Hydrocodone

0.9

--

Hydromorphone

4

20

Morphine

0.5

3

Tramadol

0.17

0.2

a For patients receiving high-dose parenteral opioids, a more conservative conversion factor should be applied (ie, lower numerical conversion factor); for example, for high-dose parenteral morphine, a conversion of 1.5 should be used for calculations instead of 3.

Conversion from fentanyl patch to extended-release oxycodone tablet: Limited data available: Children ≥11 years and Adolescents: Note: Remove fentanyl patch ≥18 hours prior to starting extended-release oxycodone. Initial dose based on current opioid regimen dose; the manufacturer suggests using the conservative conversion factor of 10 mg every 12 hours of extended-release oxycodone tablet for each 25 mcg/hour fentanyl transdermal patch; systemic assessment of this suggested conversion has not been completed, monitor patients closely.

Maintenance dose: Dosage adjustment (titration): After initiation of extended-release oxycodone tablet, adjust dose in small increments (up to 25% of current total daily dosage) no more frequently than every 1 to 2 days until desired pain control; patients may require rescue doses of an immediate-release analgesic during dose titration. Observe for signs and symptoms of opioid withdrawal or signs of oversedation/toxicity; if unacceptable adverse reactions occur, the subsequent dose may be reduced.

Discontinuation of therapy: When discontinuing chronic opioid therapy, the dose should be gradually tapered down. An optimal universal tapering schedule for all patients has not been established (Ref). Specific pediatric protocols are lacking; based on experience in adults, proposed schedules range from slow (eg, 10% reductions per week) to rapid (eg, 25% to 50% reduction every few days) (Ref). Tapering schedules should be individualized to minimize opioid withdrawal while considering patient-specific goals and concerns as well as the pharmacokinetics of the opioid being tapered. An even slower taper 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, or pausing the taper and restarting when the patient is ready (Ref). Continue to offer nonopioid analgesics as needed for pain management during the taper; consider nonopioid adjunctive treatments for withdrawal symptoms.

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

In general, oxycodone clearance may be decreased in patients with renal impairment; initiate therapy at low end of dosing range.

Immediate release: Infants, Children, and Adolescents: There are no dosage adjustments provided in the manufacturer's labeling; however, the following adjustments have been recommended (Ref):

GFR ≥50 mL/minute/1.73 m2: No dosage adjustment required.

GFR 10 to 50 mL/minute/1.73 m2: Administer 75% of dose.

GFR <10 mL/minute/1.73 m2: Administer 50% of dose.

Hemodialysis: Administer 50% of dose posthemodialysis.

Peritoneal dialysis: Administer 50% of dose.

Extended-release tablets (eg, OxyContin): Children ≥11 years and Adolescents: CrCl <60 mL/minute: Serum concentrations are increased ~50%. Initiate at the low end of the dosage range (use caution); adjust dose as clinically indicated. Doses of 33% to 50% of usual initial dosing have been recommended; if the reduced dose is less than smallest available dosage form, consider alternative analgesic.

Dosing: Hepatic Impairment: Pediatric

Immediate release: There are no dosage adjustments provided in the manufacturer's labeling; based on experience in adult patients, may consider a conservative approach of reduced initial doses; adjust dose based on clinical response.

Extended-release tablets (eg, OxyContin): Children ≥11 years and Adolescents: Initial: One-third (1/3) to one-half (1/2) of the usual starting dose; carefully titrate dose to appropriate effect. If reduced dose is less than smallest available dosage form, consider alternative analgesic.

Dosing: Adult

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

Dosage guidance:

Safety: Consider prescribing naloxone for patients with factors associated with an increased risk for overdose, such as history of overdose or substance use disorder, patients with sleep-disordered breathing, higher opioid dosages (≥50 morphine milligram equivalents [MME]/day orally [equivalent to oxycodone ≥33 mg/day]), and/or concomitant benzodiazepine use (Ref).

Dosing: Dosing provided is based on typical doses; some patients may require higher or lower doses. Individualize dosing based on patient-specific factors (eg, comorbidities, severity of pain, degree of opioid experience/tolerance) and titrate to patient-specific treatment goals (eg, improvement in function and quality of life, decrease in pain using a validated pain rating scale). Use the lowest effective dose for the shortest period of time.

Clinical considerations: Opioids may be part of a comprehensive, multimodal, patient-specific treatment plan for managing moderate to severe pain. Maximize nonopioid analgesia, if appropriate, prior to initiation of opioid analgesia (Ref).

Acute pain in opioid-naive patients

Acute pain in opioid-naive patients:

General dosing: Note: For acute non–cancer-related pain severe enough to require an opioid, utilize multimodal pain control, maximize nonopioid analgesics, and limit the quantity prescribed to the expected duration of pain severe enough to require opioids. Do not use long-acting preparations for treatment of acute pain (Ref).

Immediate release:

Oral: Initial: 5 mg every 4 to 6 hours as needed; adjust dose according to patient response. Usual dosage range: 5 to 15 mg every 4 to 6 hours as needed. If usual dose and frequency is insufficient, reassess patient and reconsider pain management strategies. For outpatient use, usually up to 20 mg/day for moderate pain or up to 30 mg/day for severe pain will suffice. Dosing is based on severity of pain and patient-specific factors; reduced dosing may be indicated in patients with comorbidities (Ref).

Rectal [Canadian product]: Usual dosage range: One (10 or 20 mg) suppository up to 3 to 4 times daily as needed. If usual dose and frequency is insufficient, reassess patient and reconsider pain management strategies (Ref).

Acute pain in patients on chronic opioid therapy

Acute pain (eg, breakthrough cancer pain) in patients on chronic opioid therapy:

Immediate release: Oral: Usual dose: In conjunction with the scheduled opioid, administer 5% to 15% (rarely up to 20%) of the 24-hour oxycodone requirement (or MME) as needed using an IR formulation every 4 to 6 hours with subsequent dosage adjustments based upon response (Ref). Note: If chronic opioid is not oxycodone, use MME calculations cautiously due to lack of complete cross-tolerance; generally, reduce calculated dose by 25% to 50%; conversions from methadone are highly variable and require extreme caution (Ref).

Acute postoperative pain, postoperative recovery/postanesthesia care unit

Acute postoperative pain, postoperative recovery/postanesthesia care unit :

Note: Optimize multimodal perioperative pain management (eg, regional or local anesthesia, nerve blocks, nonopioid analgesics, other adjuvants) to minimize opioid use (Ref). Refer to institutional protocols.

Opioid-naive patients: Immediate release:

Oral: Usual dosage range: 5 to 10 mg every 4 to 6 hours as needed. If usual dose and frequency is insufficient, reassess patient and reconsider pain management strategies (Ref). Some experts use 5 to 10 mg every 3 to 4 hours as needed (Ref).

Chronic pain, including chronic cancer pain

Chronic pain, including chronic cancer pain:

Note: Before starting opioid therapy for chronic pain, establish realistic goals for pain and function, and consider how therapy will be discontinued if benefits do not outweigh risks. Opioids, including oxycodone, 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 chronic pain from active cancer, sickle cell disease, and end-of-life care. Consider opioids, including oxycodone, only in patients who experience clinically meaningful improvement in pain and function that outweighs patient safety risks (Ref).

Opioid-naive patients: For noncancer pain, establish oxycodone requirement using IR formulations (Ref). In patients with cancer pain, may switch to a long-acting formulation earlier in the course of therapy (Ref).

Noncancer or cancer pain: Immediate release:

Oral: Initial: 2.5 to 10 mg every 4 to 6 hours as needed or scheduled around the clock (eg, cancer pain); adjust dose according to patient response (see "Titration" below). Usual maintenance dosage range: 5 to 15 mg every 4 to 6 hours as needed or scheduled around the clock. If usual dose and frequency is insufficient, reassess patient and reconsider pain management strategies. Dosing is based on severity of pain and patient-specific factors; start at the lower end of dosing range (Ref).

Titration:

Noncancer pain: Adjust dose according to patient response; if needed, increase the dose slowly in increments of no more than 25% to 50% of the total daily dose (Ref). Note: Dosages ≥50 morphine milligram equivalents (MME)/day (equivalent to oxycodone ≥33 mg/day) are likely to not have increased benefits in pain relief or function relative to overall risks. To reduce risk of overdose in noncancer pain (excluding patients with sickle cell disease and palliative care), readdress pain and reassess evidence of individual benefits and risks when increasing opioid dosage to ≥50 MME/day (equivalent to oxycodone ≥33 mg/day) (Ref).

Cancer pain: Adjust dose according to patient response; if needed, increase the fixed scheduled dose by 30% to 100% of the total dose taken in the prior 24-hour period including total amount of rescue medication used; if pain score decreased and functional assessment improved, continue current effective dosing (Ref).

Rectal [Canadian product]: Usual dosage range: One (10 or 20 mg) suppository up to 3 to 4 times daily as needed. If usual dose and frequency is insufficient, reassess patient and reconsider pain management strategies (Ref).

Opioid-tolerant patients (also refer to "Dose Conversions for Pain Management"):

Extended release:

Note: Dosage forms: Dose of ER capsules is expressed as oxycodone base and dose of ER tablets is expressed as oxycodone hydrochloride; 9 mg of oxycodone base is equivalent to 10 mg oxycodone hydrochloride. Oxycodone ER capsules and ER tablets are not bioequivalent; monitor for changes in efficacy or tolerability and adjust dose if needed. Place in therapy: Although manufacturer's labeling contains directions for initiating ER oxycodone products in patients who are opioid-naive with chronic pain, these preparations should not be used as initial therapy. Instead, initiate treatment with an IR preparation to more accurately determine the daily opioid requirement and decrease the risk of overdose. Unless pain is associated with cancer, palliative care, or sickle cell disease, reserve ER opioids for patients who have received IR opioids daily for ≥1 week yet continue to experience severe, continuous pain (Ref).

ER tablet (oxycodone hydrochloride): Oral: See "Dose Conversions for Pain Management." Calculated dose may be administered every 12 hours.

ER capsule (oxycodone base): Oral: See "Dose Conversions for Pain Management." Calculated dose may be administered every 12 hours. Maximum dose: 288 mg/day; safety of excipients in higher daily doses has not been established.

Titration: After initiation of oxycodone ER, adjust dose in increments of 25% to 50% no more frequently than every 1 to 2 days until desired pain control. Patients may require rescue doses of an IR analgesic during dose titration. Observe for signs and symptoms of opioid withdrawal or signs of oversedation/toxicity; if unacceptable adverse reactions occur, reduce the dose. Dosages ≥50 MME/day (equivalent to oxycodone ≥33 mg/day) are likely to not have increased benefits in pain relief or function relative to overall risks. To reduce risk of overdose in noncancer pain (excluding patients with sickle cell disease and palliative care), readdress pain and reassess evidence of individual benefits and risks when increasing opioid dosage to ≥50 MME/day (equivalent to oxycodone ≥33 mg/day) (Ref). Note: Some clinicians have reported better efficacy with more frequent dosing (ie, every 8 hours) (Ref); however, dosing more frequently than every 12 hours is not recommended by the manufacturer and safety has not been established.

Dose conversions for pain management:

Note: Equianalgesic conversions serve only as a general guide to estimate opioid dose equivalents for patients on scheduled doses of opioids. Multiple factors must be considered for safely individualizing conversion of opioid analgesia. In general, for noncancer pain, the decision to convert from an IR to an ER formulation should be individualized and reserved for those with severe continuous pain who have been taking opioids for ≥1 week (Ref).

Converting from IR oxycodone to ER oxycodone preparations: Total daily oral oxycodone dose may be administered in 2 divided doses. Note: Dose of ER capsules is expressed as oxycodone base and the dose of ER tablets and IR formulations is expressed as oxycodone hydrochloride; 9 mg of oxycodone base is equivalent to 10 mg oxycodone hydrochloride.

Converting between ER oxycodone preparations: Dose of ER capsules is expressed as oxycodone base and the dose of ER tablets is expressed as oxycodone hydrochloride; 9 mg of oxycodone base is equivalent to 10 mg oxycodone hydrochloride. Oxycodone ER capsules and ER tablets are not bioequivalent; monitor for changes in efficacy or tolerability and adjust dose if needed.

Converting to/from oxycodone to/from a different opioid (parenteral or oral): Refer to published equianalgesic opioid conversion data (or institutional protocols) for guidance. Conversion ratios are only approximations and substantial interpatient variability exists; therefore, it is safer to underestimate a patient’s daily oral requirement and provide breakthrough pain relief with IR formulations than to risk overestimating daily requirements. When switching to a new opioid (except to/from methadone), reduce initial daily calculated equianalgesic dose of the new opioid by 25% to 50% to adjust for lack of complete mu receptor cross-tolerance (conversions to/from methadone are highly variable and require extreme caution) (Ref).

Converting from transdermal fentanyl to oxycodone ER: Remove fentanyl patch at least 18 hours prior to starting oxycodone ER. The manufacturer suggests a conservative conversion approach of substituting each fentanyl 25 mcg/hour transdermal patch with 9 mg every 12 hours (oxycodone ER capsule) or 10 mg every 12 hours (oxycodone ER tablets). Systematic assessment of this suggested conversion has not been completed; monitor patients closely.

Converting from methadone to oxycodone ER: Extreme caution and close monitoring is required when converting methadone to another opioid. Ratio between methadone and other opioid agonists varies widely according to previous dose exposure. Methadone has a long half-life and can accumulate in the plasma.

Discontinuation or tapering of therapy:

When reducing the dose, discontinuing, or tapering long-term opioid therapy, the dose should be gradually tapered. An optimal tapering schedule has not been established. Individualize tapering based on discussions with patient to minimize withdrawal, while considering patient-specific goals and concerns and the opioid's pharmacokinetics. Proposed initial schedules range from slow (eg, 10% reduction per week or 10% reduction per month depending on duration of long-term therapy) to rapid (eg, 25% to 50% reduction every few days) (Ref). Slower tapers may be appropriate after long-term use (eg, >1 year), whereas more rapid tapers may be appropriate in patients experiencing severe adverse effects. During tapering, patients may be at an increased risk of overdose if they return to their original (or higher) opioid dose or use illicit opioids, due to rapid loss of tolerance; consider prescribing naloxone. 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 autonomic withdrawal symptoms and other adjunctive agents to treat GI symptoms and muscle spasms, as needed. Continue to offer nonopioid analgesics as needed for pain management during the taper (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.

Note: There are no specific dose adjustments provided in the manufacturer's labeling. Oxycodone is excreted as parent drug (~10%) and as active to weakly active metabolites (~47%) with varying degrees of analgesic activity (Ref); half-life is prolonged and accumulation of active metabolites occurs in patients with kidney impairment (Ref). Use of other opioids may be preferred for management of severe pain in patients with kidney impairment (Ref).

Note: When making dose adjustments for the rectal formulation [Canadian product], do not break, crush, cut, or dissolve the suppositories.

Altered kidney function:

CrCl ≥60 mL/minute: No dosage adjustment necessary.

CrCl 30 to <60 mL/minute:

Immediate release: Oral, rectal [Canadian product]: Initial: Administer 50% to 75% of usual dose no more frequently than every 6 hours (Ref). Use with caution; titrate gradually based on patient response and adverse effects.

Extended release: Oral: Initial: Administer 50% to 75% of usual dose every 12 to 24 hours; if the reduced dose is less than smallest available dosage form, consider alternative analgesic. Use with caution; titrate gradually based on patient response and adverse effects (Ref).

CrCl <30 mL/minute:

Immediate release: Oral, rectal [Canadian product]: Initial: Administer 50% of usual dose no more frequently than every 8 hours. Use with caution; titrate gradually based on patient response and adverse effects (Ref).

Extended release: Oral: ER formulations should preferably be avoided in patients with severe impairment (Ref).

Hemodialysis, intermittent (thrice weekly): Slightly dialyzable (10.6%) (Ref):

Immediate release: Oral, rectal [Canadian product]: Initial: Administer 50% of usual dose no more frequently than every 8 hours; titrate gradually based on patient response and adverse effects (Ref). Use with caution; cases of oxycodone toxicity have been reported in hemodialysis patients (Ref).

Extended release: Oral: ER formulations should preferably be avoided in patients with severe impairment (Ref).

Peritoneal dialysis:

Immediate release: Oral, rectal [Canadian product]: Initial: Administer 50% of usual dose no more frequently than every 8 hours; titrate gradually based on patient response and adverse effects (Ref); use with caution.

Extended release: Oral: ER formulations should preferably be avoided in patients with severe impairment (Ref).

CRRT: Drug clearance is dependent on the effluent flow rate, filter type, and method of renal replacement. Recommendations are based on high-flux dialyzers and effluent flow rates of 20 to 25 mL/kg/hour (or ~1,500 to 3,000 mL/hour) unless otherwise noted. Close monitoring of response (analgesia) and adverse reactions (eg, sedation, CNS and respiratory depression) due to drug accumulation is important.

Immediate release: Oral, rectal [Canadian product]: Initial: Administer 50% of usual dose no more frequently than every 6 hours; titrate gradually based on patient response and adverse effects (Ref).

Extended release: Oral: Avoid use.

PIRRT (eg, sustained, low-efficiency diafiltration): Drug clearance is dependent on the effluent flow rate, filter type, and method of renal replacement. Close monitoring of response (analgesia) and adverse reactions (eg, sedation, CNS and respiratory depression) due to drug accumulation is important.

Immediate release: Oral, rectal [Canadian product]: Initial: Administer 50% of usual dose no more frequently than every 6 hours; titrate gradually based on patient response and adverse effects (Ref).

Extended release: Oral: Avoid use.

Dosing: Hepatic Impairment: Adult

Immediate release: Initiate therapy at 33% to 50% the usual dosage and titrate carefully. For patients with severe impairment, consider extending the dosing interval based on response and tolerability (eg, every 6 to 12 hours) (Ref).

Extended release tablets or Extended release capsules: Initial: Initiate oxycodone ER with 33% to 50% of the calculated recommended dose. If reduced dose is less than smallest available dosage form consider alternative analgesic.

Adverse Reactions (Significant): Considerations
Opioid-induced constipation

Opioid-induced constipation (OIC) is the most common subtype of opioid-induced bowel dysfunction, which is a broader term that encompasses additional GI opioid-induced adverse reactions including nausea, vomiting, and gastroesophageal reflux. Symptoms of OIC may include decreased frequency of bowel movements, straining to pass bowel movements, a sense of incomplete evacuation, and/or hard stools (Ref). Tolerance does not develop to OIC (Müller-Lissner 2017, Rossi 2019). Symptoms are reversible after discontinuation of the opioid (Ref). OIC is the most common reason for discontinuation reported by patients and may often result in a longer hospital stay and increased overall healthcare costs (Ref). Opioid dose or route of administration does not appear to alter risk (Ref).

Mechanism: Time-related; mu-opioid receptor stimulation in the GI tract results in delayed gastric emptying, decreased peristalsis, decreased water and chloride secretion into the intestinal lumen, and slowed bowel motility (Ref).

Onset: Varied; changes in peristalsis may occur 5 to 25 minutes after administration of opioids (Ref). However, OIC is defined based on a 7-day period of change (Ref).

Risk factors:

• Increased dosing frequency (ie, daily vs 2 to 3 times per week) (Ref)

• Chronic opioid administration (Ref)

• Concurrent use of other medications that cause constipation

Opioid-induced neurotoxicity

Opioid-induced neurotoxicity is an opioid-related adverse drug reaction more commonly associated with opioids that have active metabolites (eg, codeine, hydromorphone, meperidine, morphine, oxycodone). Symptoms may include allodynia, delirium, hallucinations, hyperalgesia, hypersomnolence, myoclonus, tremor, and seizures (Ref).

Mechanism: Not clearly established; hypothesized to result from an accumulation of neuroexcitatory opioid metabolites (Ref).

Risk factors:

• High opioid dose (Ref)

• Dehydration (Ref)

• Kidney failure (Ref)

• Infection (Ref)

• Advanced age (Ref)

Opioid-induced respiratory depression

Serious, life-threatening, or fatal opioid-induced respiratory depression (OIRD) may occur with use of oxycodone in adult and pediatric patients. Effects include hypoventilation, hypoxia, hypercapnia, and respiratory acidosis, as well as reduced ventilatory responses to hypoxia and hypercapnia (Ref).

Mechanism: Dose-related; stimulation of mu-opioid receptors in the brainstem leads to suppression of the respiratory control network and depression of normal hypoxic and hypercapnic ventilatory responses (Ref).

Onset: Rapid; OIRD reported within 2 hours after administration of oxycodone IR capsule (Ref).

Risk factors:

• Higher doses of intraoperative opioids (eg, morphine-equivalent daily dose ~40 mg in opioid-naïve patients) (Ref)

• Initiation of therapy or dose increase

• Opioid naïve (Ref)

• Opioid misuse (Ref)

• Acute overdose

• Concurrent administration of benzodiazepines, alcohol, or other CNS depressants

• Postoperative patients with comorbid cardiac or respiratory disease (Ref)

• Age >60 years (Ref)

• Males (Ref)

• Sleep disorder (eg, obstructive sleep apnea) (Ref)

• Chronic pulmonary disease

• Cachexia

• Debilitation

Opioid-induced withdrawal

Abruptly stopping or reducing opioid use in patients with physical dependence on opioids can precipitate opioid-induced withdrawal (OIW) in adult and pediatric patients (Ref). Physical symptoms may include nausea, vomiting, diarrhea, abdominal cramps, tachycardia, chills, muscle aches, bone pain, agitation, anxiety, and insomnia. Psychosis has also been reported in case reports and case series (Ref). The presence and severity of withdrawal symptoms are associated with an increased risk of nonfatal overdose and receptive syringe sharing in people who inject drugs (Ref). Avoidance of withdrawal symptoms may drive continued opioid use (Kosten 2019). Symptoms typically dissipate over 4 to 7 days (Ref).

Mechanism: Withdrawal; opioids bind to mu-opioid receptors on neurons in the locus coeruleus (LC) of the brainstem, causing decreased norepinephrine (NE) release. Upon abrupt discontinuation, the absence of opioid stimulation causes LC hyperactivity, excessive NE release, and subsequent autonomic hyperactivity (Ref).

Onset: Rapid; symptoms typically occur within 12 hours of discontinuation and peak at 36 to 72 hours; varies based on half-life of opioid (Ref).

Risk factors:

• Higher cumulative dose (Ref)

• Prolonged exposure to opioids (Ref)

• Initiation of mixed agonist/antagonist or partial agonist analgesics (eg, buprenorphine, butorphanol, nalbuphine, pentazocine) (Ref) in patients currently or recently taking full mu-opioid agonists may cause precipitated opioid withdrawal. (Precipitated withdrawal is similar to OIW, but with a faster onset (Ref).

• Opioid use disorder (Ref)

• Abrupt discontinuation or dose reduction (Ref)

• Concurrent use of benzodiazepines (Ref) or antipsychotics (Ref)

• Age <6 years, especially infants <6 months of age (Ref)

• Preexisting cognitive impairment (Ref)

• Critical illness involving CNS (Ref)

Adverse Reactions

The following adverse drug reactions and incidences are derived from product labeling unless otherwise specified. Adverse reactions for the immediate release (IR) product reported in adults and in adolescents and adults for the extended release (ER) product unless otherwise indicated.

>10%:

Dermatologic: Pruritus (ER: 3% to 13%; IR: ≥3%)

Gastrointestinal: Constipation (ER: 5% to 23%; IR: ≥3%) (table 1), nausea (ER: 11% to 23%; IR: ≥3%), vomiting (ER: 4% to 21%; IR: ≥3%)

Oxycodone: Adverse Reaction: Constipation

Drug (Oxycodone)

Placebo

Population

Dosage Form

Indication

Number of Patients (Oxycodone)

Number of Patients (Placebo)

9%

N/A

Adolescents

Extended-release tablets

N/A

140

N/A

5%

0.5%

N/A

Extended-release capsules

Moderate to severe chronic lower back pain

193

196

23%

7%

N/A

Extended-release tablets

N/A

227

45

≥3%

N/A

N/A

Immediate-release tablets

N/A

191

N/A

Nervous system: Dizziness (ER: 2% to 13%; IR: ≥3%), drowsiness (ER [adults]: 9% to 23%; ER [adolescents]: 1% to 5%; IR: ≥3%), headache (ER: 14%; IR: ≥3%)

Miscellaneous: Fever (ER: 1% to 11%; IR: ≥3%)

1% to 10%:

Cardiovascular: Deep vein thrombosis (IR: <3%), edema (≤5%), flushing (ER [adults]: 1% to 5%), heart failure (IR: <3%), hypertension (ER [adults]: 1% to 5%), hypotension (IR: <3%), orthostatic hypotension (ER: 1% to 5%), palpitations (IR: <3%), peripheral edema (<3%), tachycardia (≤5%), thrombophlebitis (IR: <3%), vasodilation (IR: <3%)

Dermatologic: Diaphoresis (≤5%), excoriation of skin (ER [adults]: 1% to 5%), hyperhidrosis (≤5%), skin photosensitivity (IR: <3%), skin rash (≤5%), urticaria (IR: <3%)

Endocrine & metabolic: Hyperglycemia (≤5%), hypochloremia (ER [adolescents]: 1% to 5%), hyponatremia (ER [adolescents]: 1% to 5%), weight loss (ER [adolescents]: 1% to 5%)

Gastrointestinal: Abdominal pain (≤5%, including upper abdominal pain), anorexia (≤5%), decreased appetite (≤5%), diarrhea (≤6%), dyspepsia (≤5%), dysphagia (<3%), gastritis (ER: 1% to 5%), gastroesophageal reflux disease (ER: 1% to 5%), gingivitis (IR: <3%), glossitis (IR: <3%), hiccups (ER: 1% to 5%), xerostomia (≤6%)

Genitourinary: Dysuria (ER [adolescents]: 1% to 5%; ER [adults]: <1%), urinary retention (ER [adolescents]: 1% to 5%; ER [adults]: <1%), urinary tract infection (IR: <3%)

Hematologic & oncologic: Anemia (IR: <3%, including iron deficiency anemia), decreased platelet count (ER [adolescents]: 1% to 5%), decreased red blood cells (ER [adolescents]: 1% to 5%), febrile neutropenia (ER [adolescents]: 1% to 5%), hemorrhage (IR: <3%), leukopenia (IR: <3%), neutropenia (ER [adolescents]: 1% to 5%)

Hepatic: Increased serum alanine aminotransferase (ER [adolescents]: 1% to 5%)

Hypersensitivity: Hypersensitivity reaction (IR: <3%)

Infection: Herpes simplex infection (IR: <3%), infection (IR: <3%), sepsis (IR: <3%)

Nervous system: Abnormal dreams (ER: ≤5%), agitation (≤5%), anxiety (≤5%), asthenia (1% to 6%), changes in thinking (ER: 1% to 5%), chills (≤5%), confusion (≤5%), depression (ER [adolescents]: 1% to 5%; ER [adults]: <1%), dysphoria (ER: 1% to 5%), euphoria (ER: ≤5%), fatigue (ER: 1% to 5%), hypertonia (IR: <3%), hypoesthesia (≤5%), insomnia (ER: 1% to 5%; IR: ≥3%), irritability (ER [adults]: 1% to 5%), lethargy (ER: 1% to 5%), migraine (adults: ≤5%), nervousness (≤5%), neuralgia (IR: <3%), paresthesia (ER [adolescents]: 1% to 5%; ER [adults]: <1%), personality disorder (IR: <3%), tremor (adults: ≤5%), twitching (ER: 1% to 5%), withdrawal syndrome (ER [adults]: ≤5%)

Neuromuscular & skeletal: Arthralgia (adults: ≤5%), arthritis (IR: <3%), back pain (adults: ≤5%), gout (IR: <3%), laryngospasm (IR: <3%), limb pain (ER [adolescents]: 1% to 5%), musculoskeletal pain (ER: ≤5%), myalgia (≤5%), neck pain (IR: <3%), ostealgia (IR: <3%), pathological fracture (IR: <3%)

Ophthalmic: Amblyopia (IR: <3%), blurred vision (ER [adults]: 1% to 5%)

Respiratory: Bronchitis (IR: <3%), cough (adults: ≤5%), dyspnea (≤5%), epistaxis (IR: <3%), flu-like symptoms (IR: <3%), laryngismus (IR: <3%), oropharyngeal pain (ER: 1% to 5%), oxygen saturation decreased (ER [adolescents]: 1% to 5%), pharyngitis (IR: <3%), rhinitis (IR: <3%), sinusitis (IR: <3%)

Miscellaneous: Accidental injury (<3%), seroma (ER [adolescents]: 1% to 5%)

<1% (any formulation or population):

Cardiovascular: Chest pain, depression of ST segment on ECG, syncope

Dermatologic: Exfoliative dermatitis, night sweats, xeroderma

Endocrine & metabolic: Dehydration, increased thirst

Gastrointestinal: Dysgeusia, eructation, flatulence, increased appetite, stomatitis

Genitourinary: Erectile dysfunction, hematuria, polyuria

Hematologic & oncologic: Lymphadenopathy

Hepatic: Increased gamma-glutamyl transferase

Hypersensitivity: Facial edema

Nervous system: Abnormal gait, amnesia, depersonalization, emotional lability, hallucination, hypotonia, malaise, memory impairment, restlessness, sleep disturbance, speech disturbance, stupor, vertigo, voice disorder

Neuromuscular & skeletal: Hyperkinetic muscle activity

Ophthalmic: Visual disturbance

Otic: Tinnitus

Frequency not defined (any formulation or population):

Cardiovascular: Circulatory depression, shock

Respiratory: Respiratory depression

Postmarketing (any formulation or population):

Endocrine & metabolic: Amenorrhea, SIADH

Gastrointestinal: Cholestasis, dental caries, diverticulitis of the gastrointestinal tract (exacerbation), intestinal obstruction

Genitourinary: Hypogonadism

Hepatic: Increased liver enzymes

Nervous system: Aggressive behavior, allodynia (opioid-induced hyperalgesia) (FDA Safety Communication 2023), drug abuse, drug dependence, hyperalgesia, mood changes, neonatal withdrawal, seizure, suicidal ideation, suicidal tendencies

Respiratory: Pharyngeal edema

Contraindications

Hypersensitivity (eg, anaphylaxis, angioedema) to oxycodone or any component of the formulation; significant respiratory depression; hypercapnia; acute or severe bronchial asthma in an unmonitored setting or in the absence of resuscitative equipment; GI obstruction, including paralytic ileus (known or suspected).

Canadian labeling: Additional contraindications (not in US labeling): Hypersensitivity to other opioids; suspected surgical abdomen (eg, acute appendicitis or pancreatitis); any disease/condition that affects bowel transit; mild pain that can be managed with other pain medications (immediate release, suppository); mild, intermittent or short duration pain that can be managed with other pain medications or acute pain (extended release); chronic obstructive airway; status asthmaticus; cor pulmonale; acute alcoholism; delirium tremens; convulsive disorders; severe CNS depression; increased cerebrospinal or intracranial pressure; head injury; monoamine oxidase (MAO) inhibitors (concomitant use or within 14 days of therapy); pregnant women or during labor and delivery; breastfeeding.

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

Warnings/Precautions

Concerns related to adverse effects:

• CNS depression: May cause CNS depression, which may impair physical or mental abilities; patients must be cautioned about performing tasks that require mental alertness (eg, operating machinery, driving).

• Hypotension: May cause severe hypotension (including orthostatic hypotension and syncope); use with caution in patients with hypovolemia, cardiovascular disease (including acute 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.

• Phenanthrene hypersensitivity: Use with caution in patients with hypersensitivity reactions to other phenanthrene-derivative opioid agonists (codeine, hydrocodone, hydromorphone, levorphanol, oxymorphone).

• Respiratory depression: Carbon dioxide retention from opioid-induced respiratory depression can exacerbate the sedating effects of opioids. Patients and caregivers should be educated on how to recognize respiratory depression and the importance of getting emergency assistance immediately (eg, calling 911) in the event of known or suspected overdose.

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 adrenocortical insufficiency, including Addison disease; dose adjustment may be required. Long-term opioid use may cause secondary hypogonadism, which may lead to mood disorders and osteoporosis (Brennan 2013).

• Biliary tract impairment: Use with caution in patients with biliary tract dysfunction, including acute pancreatitis; may cause constriction of 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.

• 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 in patients with hepatic impairment; oxycodone clearance may decrease.

• 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 potential increased risk for opioid use disorder and overdose; more frequent monitoring is recommended (CDC [Dowell 2022]).

• 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; dose adjustment may be required.

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

• Renal impairment: Use with caution in patients with renal impairment; oxycodone clearance may decrease.

• 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.

• Seizures: Use with caution in patients with a history of seizure disorders; may cause or exacerbate preexisting seizures.

• Sleep-related disorders: Use with caution in patients with sleep-related disorders, including sleep apnea, due to increased risk for respiratory and CNS depression. Monitor carefully and titrate dosage cautiously in patients with mild sleep-disordered breathing. Avoid opioids in patients with moderate to severe sleep-disordered breathing (CDC [Dowell 2022]).

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

Special populations:

• Older adult: Use opioids with caution in older adults; may be more sensitive to adverse effects. Clearance may also be reduced in older adults (with or without renal impairment) resulting in a narrow therapeutic window and increased adverse effects. Monitor closely for adverse effects associated with opioid therapy (eg, respiratory and CNS depression, falls, cognitive impairment, constipation) (CDC [Dowell 2022]). Consider the use of alternative nonopioid analgesics in these patients when possible.

Dosage form specific issues:

• Benzyl alcohol and derivatives: Some dosage forms may contain sodium benzoate/benzoic acid; benzoic acid (benzoate) is a metabolite of benzyl alcohol; large amounts of benzyl alcohol (≥99 mg/kg/day) have been associated with a potentially fatal toxicity (“gasping syndrome”) in neonates; the “gasping syndrome” consists of metabolic acidosis, respiratory distress, gasping respirations, CNS dysfunction (including convulsions, intracranial hemorrhage), hypotension, and cardiovascular collapse (AAP ["Inactive" 1997]; CDC 1982); some data suggest that benzoate displaces bilirubin from protein-binding sites (Ahlfors 2001); avoid or use dosage forms containing benzyl alcohol derivative with caution in neonates. See manufacturer's labeling.

• Extended-release tablets: Tablets may be difficult to swallow and could become lodged in throat; patients with swallowing difficulties may be at increased risk. Cases of intestinal obstruction or diverticulitis exacerbation have also been reported, including cases requiring medical intervention to remove the tablet; patients with an underlying GI disease (eg, esophageal cancer, colon cancer) may be at increased risk.

Other warnings/precautions:

• Abuse/misuse/diversion: Use with caution in patients with a history of substance abuse disorder; potential for drug dependency exists. Other factors associated with increased risk for misuse include concomitant depression or other mental health conditions, higher opioid dosages, or taking other CNS depressants. Consider offering naloxone prescriptions in patients with an increased risk for overdose, such as history of overdose or substance use disorder, higher opioid dosages (≥50 morphine milligram equivalents [MME]/day orally), concomitant benzodiazepine use, and patients at risk for returning to a high dose after losing tolerance (CDC [Dowell 2022]).

• Appropriate use: Outpatient setting: Opioids should not be used as first-line therapy for acute (<1-month duration), subacute (1- to 3-month duration), or chronic pain (>3-month duration [outside of end-of-life or palliative care, active cancer treatment, sickle cell disease, or medication-based opioid use disorder treatment]). Preferred management includes nonpharmacologic therapy and non-opioid therapy (eg, nonsteroidal anti-inflammatory drugs, acetaminophen, certain antiseizure medications and antidepressants) as appropriate for the specific condition. If opioid therapy is initiated, it should be combined with nonpharmacologic and non-opioid therapy, as appropriate. Prior to initiation, known risks and realistic benefits of opioid therapy should be discussed with the patient. Therapy should be initiated at the lowest effective dosage using IR opioids (instead of ER/long-acting opioids). For the treatment of acute pain, therapy should only be given for the expected duration of pain severe enough to require opioids and prescribed as needed (not scheduled). For the treatment of subacute and chronic pain, 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. Risk to patients increases with higher opioid dosages. Dosages ≥50 MME/day are likely to not have increased benefit to pain relief or function relative to overall risk to patients; before increasing dosage to ≥50 MME/day, readdress pain and reassess evidence of individual benefits and risks (CDC [Dowell 2022]).

• 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), or have experienced opioid-induced respiratory depression/opioid overdose. 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.

• REMS program: To ensure that the benefits of opioid analgesics outweigh the risks of substance use disorder, abuse, and misuse, a REMS is required. Drug companies with approved opioid analgesic products must make REMS-compliant education programs available to health care providers. Health care providers are 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.

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

Dosage Forms Considerations

Xtampza ER: Strength is expressed in terms of oxycodone base.

9 mg equivalent to 10 mg oxycodone hydrochloride

13.5 mg equivalent to 15 mg oxycodone hydrochloride

18 mg equivalent to 20 mg oxycodone hydrochloride

27 mg equivalent to 30 mg oxycodone hydrochloride

36 mg equivalent to 40 mg oxycodone hydrochloride

Dosage Forms: US

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

Capsule, Oral, as hydrochloride:

Generic: 5 mg

Capsule ER 12 Hour Abuse-Deterrent, Oral:

Xtampza ER: 9 mg (100 ea); 13.5 mg (100 ea); 18 mg (100 ea); 27 mg (100 ea); 36 mg (100 ea)

Concentrate, Oral, as hydrochloride:

Generic: 100 mg/5 mL (30 mL)

Solution, Oral, as hydrochloride:

Generic: 5 mg/5 mL (5 mL, 15 mL, 473 mL, 500 mL)

Tablet, Oral, as hydrochloride:

Oxaydo: 5 mg, 7.5 mg

Roxicodone: 5 mg [DSC] [scored]

Roxicodone: 15 mg [scored; contains fd&c blue #2 (indigotine,indigo carmine), quinoline yellow (d&c yellow #10)]

Roxicodone: 30 mg [scored]

Generic: 5 mg, 10 mg, 15 mg, 20 mg, 30 mg

Tablet Abuse-Deterrent, Oral:

RoxyBond: 15 mg, 30 mg [contains fd&c blue #2 (indigotine,indigo carmine)]

Tablet Abuse-Deterrent, Oral, as hydrochloride:

RoxyBond: 5 mg

Tablet ER 12 Hour Abuse-Deterrent, Oral, as hydrochloride:

OxyCONTIN: 10 mg, 15 mg, 20 mg, 30 mg, 40 mg, 60 mg

OxyCONTIN: 80 mg [contains fd&c blue #2 (indigo carm) aluminum lake]

Generic: 10 mg, 15 mg [DSC], 20 mg, 30 mg [DSC], 40 mg, 60 mg [DSC], 80 mg

Generic Equivalent Available: US

May be product dependent

Pricing: US

Capsule ER 12 Hour Abuse-Deterrent (Xtampza ER Oral)

9 mg (per each): $7.65

13.5 mg (per each): $11.26

18 mg (per each): $14.28

27 mg (per each): $19.86

36 mg (per each): $24.44

Capsules (oxyCODONE HCl Oral)

5 mg (per each): $1.84

Concentrate (oxyCODONE HCl Oral)

100 mg/5 mL (per mL): $5.60 - $12.33

Solution (oxyCODONE HCl Oral)

5 mg/5 mL (per mL): $1.05 - $1.60

Tablet Abuse-Deterrent (RoxyBond Oral)

5 mg (per each): $15.28

15 mg (per each): $18.84

30 mg (per each): $25.40

Tablet ER 12 Hour Abuse-Deterrent (oxyCODONE HCl ER Oral)

10 mg (per each): $4.23 - $5.03

20 mg (per each): $7.88 - $9.37

40 mg (per each): $13.10 - $16.05

80 mg (per each): $28.01

Tablet ER 12 Hour Abuse-Deterrent (OxyCONTIN Oral)

10 mg (per each): $6.57

15 mg (per each): $9.66

20 mg (per each): $12.24

30 mg (per each): $17.03

40 mg (per each): $20.95

60 mg (per each): $29.68

80 mg (per each): $36.58

Tablets (Oxaydo Oral)

5 mg (per each): $12.86

7.5 mg (per each): $19.26

Tablets (oxyCODONE HCl Oral)

5 mg (per each): $0.08 - $0.62

10 mg (per each): $0.15 - $1.45

15 mg (per each): $0.12 - $2.37

20 mg (per each): $0.27 - $2.32

30 mg (per each): $0.18 - $4.49

Tablets (Roxicodone Oral)

15 mg (per each): $6.23

30 mg (per each): $12.23

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.

Dosage Forms: Canada

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

Suppository, Rectal:

Supeudol 10: 10 mg (12 ea)

Supeudol 20: 20 mg (12 ea)

Tablet, Oral, as hydrochloride:

Oxy-IR: 5 mg, 10 mg, 20 mg

Supeudol: 5 mg, 10 mg, 20 mg

Generic: 5 mg, 10 mg, 20 mg

Tablet ER 12 Hour Abuse-Deterrent, Oral, as hydrochloride:

OxyNEO: 10 mg, 15 mg, 20 mg, 30 mg, 40 mg, 60 mg

OxyNEO: 80 mg [contains fd&c blue #2 (indigo carm) aluminum lake]

Tablet Extended Release 12 Hour, Oral:

Generic: 5 mg, 10 mg, 15 mg, 20 mg, 30 mg, 40 mg, 60 mg, 80 mg

Additional Information

OxyContin tablets deliver medication over 12 hours; release is pH independent. Equianalgesic doses: oral oxycodone 30 mg = morphine 10 mg IM = single oral dose morphine 60 mg or chronic dosing oral morphine 30 mg. Note: Not appropriate for OxyContin initial dose calculation.

Oxaydo utilizes Acura Pharmaceutical's Aversion technology which may help discourage misuse and abuse potential. Reduced abuse potential of Oxaydo compared to other immediate-release oxycodone tablet formulations has not been proven; the FDA has required a post-approval epidemiological study to determine whether the formulation actually results in a decrease of misuse/abuse. In one clinical trial in nondependent recreational opioid users, the "drug-liking" responses and safety of crushed Oxaydo tablets were compared to crushed immediate-release oxycodone tablets following the self-administered intranasal use. A small difference in "drug-liking" scores was observed, with lower scores reported in the crushed Oxaydo group. In regard to safety, there was an increased incidence of nasopharyngeal and facial adverse events in the Oxaydo group. In addition, there was decreased ability in the Oxaydo group to completely administer the 2 crushed Oxaydo tablets intranasally within a set time period. However, whether these differences translate into a significant clinical difference is unknown. Of note, pharmacokinetic studies showed that Oxaydo is bioequivalent with oxycodone immediate-release tablets with no differences in Tmax and half-life when administered in the fasted state.

Controlled Substance

C-II

Administration: Pediatric

Oral:

Immediate release (capsule, oral solution, tablets): May administer with food to decrease GI upset:

Oral solution: Available in 2 strengths: 1 mg/mL and a concentrated oral solution (20 mg/mL). Precautions should be taken to avoid confusion between the different concentrations; prescriptions should have the concentration specified as well as the dose clearly represented as milligram (mg) of oxycodone, not volume (mL). The enclosed calibrated oral syringe should always be used to administer the concentrated oral solution to ensure the dose is measured and administered accurately. The concentrated oral solution (20 mg/mL) should only be used in opioid-tolerant patients (taking ≥30 mg/day of oxycodone or equivalent for ≥1 week).

Tablet (Oxaydo): Swallow whole with adequate water to ensure complete swallowing immediately after placing in the mouth; the formulation uses technology designed to discourage common methods of tampering to prevent misuse/abuse. The tablet should not be wet prior to placing in the mouth. Do not crush, chew, or dissolve nor administer via feeding tubes (eg, gastric, NG) due to potential for obstruction.

Extended release: Tablet (eg, OxyContin): May administer with food to decrease GI upset. Swallow whole; do not moisten, dissolve, cut, crush, chew, or break as this would result in rapid release of oxycodone and absorption of a potentially fatal dose of drug. Administer one at a time and follow each with water immediately after placing in the mouth. For oral use only; do not administer rectally; increased risk of adverse events due to better rectal absorption.

Administration: Adult

Appropriate laxatives should be administered to avoid the constipating side effects associated with use. Antiemetics may be needed for persistent nausea. Some dosage forms (eg, ER) may not be appropriate for administration through feeding tubes (eg, gastric, NG). Refer to product labeling.

ER dosage forms:

Oral:

Tablet: Administer with or without food. Swallow tablet whole. Do not moisten, dissolve, cut, crush, break, or chew extended release tablets. Extended release tablets should be administered one at a time and each followed with water immediately after placing in the mouth.

Capsule: Administer each dose with food and approximately the same amount. For patients with difficulty swallowing, capsule may be opened and the contents sprinkled on soft foods (eg, applesauce, pudding, yogurt, ice cream, jam) or into a cup for administration directly into the mouth. Rinse mouth immediately afterwards to ensure all contents have been swallowed. Contents of capsule may also be administered through a nasogastric (NG) tube or gastrostomy tube (G-tube). Flush tube with water first, then pour capsule contents directly into tube (do not premix capsule contents with fluid that will be used to flush them through the tube). After contents have been placed in tube, flush tube with 15 mL of water, milk, or liquid nutritional supplement once and then repeat twice with 10 mL.

Bariatric surgery: Tablet, abuse deterrent and extended or controlled release: Some institutions may have specific protocols that conflict with these recommendations; refer to institutional protocols as appropriate. Roxybond (oxycodone) tablets are formulated to have increased resistance to cutting, crushing, grinding, or breaking. Pharmacokinetics are similar to other IR oxycodone formulations. Nonabuse-deterrent IR tablet, capsule, and oral solution formulations are available. Oxycodone ER capsule contents may be taken by sprinkling the contents onto soft food (ie, applesauce, ice cream, yogurt). If safety and efficacy can be effectively monitored, no change in formulation or administration is required after bariatric surgery; however, clinicians should be advised that oral morphine has been shown to have significantly increased Cmax and decreased Tmax in the immediate (1 to 2 weeks) and long-term (6-months) period after bariatric surgery.

IR dosage forms:

Oral:

Capsule: Administer with or without food.

Oral solution: Administer with or without food. Available in two strengths; 1 mg/mL and a concentrated oral solution (20 mg/mL). Precautions should be taken to avoid confusion between the different concentrations; prescriptions should have the concentration specified as well as the dose clearly represented as milligram (mg) of oxycodone, not volume (mL). The enclosed calibrated oral syringe should always be used to administer the concentrated oral solution to ensure the dose is measured and administered accurately. The concentrated oral solution (20 mg/mL) may be used in opioid-tolerant patients (taking ≥30 mg/day of oxycodone or equivalent for ≥1 week); may also be used in palliative care patients who have difficulty and/or are unable to swallow. The concentrate is not substantially absorbed sublingually/orally and requires the GI tract for effective absorption (Ref).

Tablets:

Without abuse deterrent: Administer with or without food. When administered with food, onset may be delayed.

With abuse deterrent:

Oxaydo: Administer with or without food. Do not crush, chew, or dissolve the tablets. Due to inactive ingredient that causes nasal burning (upon snorting) and throat irritation, the tablet must be swallowed whole with enough water to ensure complete swallowing immediately after placing in the mouth. The tablet should not be wet prior to placing in the mouth.

Rectal:

Suppository [Canadian product]: Administer rectally. Do not break, crush, cut, or dissolve the suppositories.

Storage/Stability

Store at 25°C (77°F); excursions permitted between 15°C to 30°C (59°F to 86°F). Protect from light and moisture.

Suppository [Canadian product]: Store below 25°C (77°F).

Use

Immediate-release formulations (capsules, oral solution, and tablets [eg, Oxaydo, RoxyBond, Roxicodone]): Management of acute or chronic moderate to severe pain for which the use of an opioid analgesic is appropriate and alternative treatments are inadequate (FDA approved in adults).

Extended-release capsule (Xtampza ER): Management of pain severe enough to require daily, around-the-clock, long-term opioid treatment and for which alternative treatment options are inadequate (FDA approved in adults).

Extended-release tablets (eg, OxyContin): Management of pain severe enough to require daily, around-the-clock, long-term opioid therapy for which alternative treatment options are inadequate in adults and also opioid-tolerant in pediatric patients (FDA approved in pediatric patients ≥11 years receiving and tolerating ≥20 mg/day oxycodone or its equivalent and adults).

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

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

OxyCODONE may be confused with HYDROcodone, oxyBUTYnin, OxyCONTIN, oxyMORphone

OxyCONTIN may be confused with MS Contin, oxyBUTYnin, oxyCODONE, oxyMORphone, oxytocin

OxyFast may be confused with Roxanol

Roxicodone may be confused with Roxanol

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 patient harm when used in error.

Metabolism/Transport Effects

Substrate of CYP2D6 (minor), CYP3A4 (major); Note: Assignment of Major/Minor substrate status based on clinically relevant drug interaction potential

Drug Interactions

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

Alizapride: May enhance the CNS depressant effect of CNS Depressants. 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

Amphetamines: May enhance the analgesic effect of Opioid Agonists. 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

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

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

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

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

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 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

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

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

CYP3A4 Inhibitors (Moderate): May increase the serum concentration of OxyCODONE. Serum concentrations of the active metabolite Oxymorphone may also be increased. Risk C: Monitor therapy

CYP3A4 Inhibitors (Strong): May enhance the adverse/toxic effect of OxyCODONE. CYP3A4 Inhibitors (Strong) may increase the serum concentration of OxyCODONE. Serum concentrations of the active metabolite oxymorphone may also be increased. Risk C: Monitor therapy

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

Monoamine Oxidase Inhibitors: OxyCODONE may enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Management: Use of oxycodone is not recommended for patients taking MAOIs or within 14 days of MAOI discontinuation. If combined, use test doses and frequent titration of small doses while monitoring blood pressure, CNS depression, and signs of serotonin syndrome. Risk D: Consider therapy modification

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

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: Opioid Agonists (metabolized by CYP3A4) may enhance the serotonergic effect of Nefazodone. This could result in serotonin syndrome. Nefazodone may increase the serum concentration of Opioid Agonists (metabolized by CYP3A4). Management: If concomitant use of opioid agonists that are metabolized by CYP3A4 and nefazodone is necessary, consider dose reduction of the opioid until stable drug effects are achieved. Monitor for increased opioid effects and serotonin syndrome/serotonin toxicity. Risk D: Consider therapy modification

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

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

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

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

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

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

Procarbazine: May enhance the CNS depressant effect of CNS Depressants. 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

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

Serotonergic Agents (High Risk): Opioid Agonists (metabolized by CYP3A4) 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

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

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

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

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

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

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

Zuranolone: May enhance the CNS depressant effect of CNS Depressants. Management: Consider alternatives to the use of zuranolone with other CNS depressants or alcohol. If combined, consider a zuranolone dose reduction and monitor patients closely for increased CNS depressant effects. Risk D: Consider therapy modification

Dietary Considerations

Instruct patient to avoid high-fat meals when taking some products (food has no effect on the reformulated OxyContin).

Reproductive Considerations

Chronic opioid use may cause hypogonadism and hyperprolactinemia which may decrease fertility in patients of reproductive potential. Menstrual cycle disorders (including amenorrhea), erectile dysfunction, and impotence have been reported. The incidence of hypogonadism may be increased with the use of opioids in high doses or long-acting opioid formulations. It is not known if the effects on fertility are reversible. Monitor patients on long-term therapy (de Vries 2020; Gadelha 2022).

Consider family planning, contraception, and the effects on fertility prior to prescribing opioids for chronic pain to patients who could become pregnant (ACOG 2017; CDC [Dowell 2022]).

Pregnancy Considerations

Oxycodone crosses the placenta (Kokki 2012).

Maternal use of opioids may be associated with poor fetal growth, stillbirth, and preterm delivery (CDC [Dowell 2022]). Opioids used as part of obstetric analgesia/anesthesia during labor and delivery may temporarily affect the fetal heart rate (ACOG 2019).

Neonatal abstinence syndrome (NAS)/neonatal opioid withdrawal syndrome (NOWS) may occur following prolonged in utero exposure to opioids (CDC [Dowell 2022]). NAS/NOWS may be life-threatening if not recognized and treated and requires management according to protocols developed by neonatology experts. Presentation of symptoms varies by opioid characteristics (eg, immediate release, sustained release), time of last dose prior to delivery, drug metabolism (maternal, placental, and infant), net placental transfer, as well as other factors (AAP [Hudak 2012]; AAP [Patrick 2020]). Clinical signs characteristic of withdrawal following in utero opioid exposure include excessive crying or easily irritable, fragmented sleep (<2 to 3 hours after feeding), tremors, increased muscle tone, or GI dysfunction (hyperphagia, poor feeding, feeding intolerance, watery or loose stools) (Jilani 2022). NAS/NOWS occurs following chronic opioid exposure and would not be expected following the use of opioids at delivery (AAP [Patrick 2020]).

Monitor infants of mothers on long-term/chronic opioid therapy for symptoms of withdrawal. Symptom onset reflects the half-life of the opioid used. Monitor infants for at least 3 days following exposure to immediate-release opioids; monitor for at least 4 to 7 days following exposure to sustained-release opioids (AAP [Patrick 2020]; CDC [Dowell 2022]). Monitor newborns for excess sedation and respiratory depression when opioids are used during labor.

When opioids are needed to treat acute pain in pregnant patients, the lowest effective dose for only the expected duration of pain should be prescribed (CDC [Dowell 2022]).

Opioid use for pain following vaginal or cesarean delivery should be made as part of a shared decision-making process. A stepwise, multimodal approach to managing postpartum pain is recommended. A low-dose, low-potency, short-acting opioid such as oxycodone can be used to treat acute pain associated with delivery when needed (ACOG 2021).

Opioids are not preferred for the treatment of chronic noncancer pain during pregnancy; consider strategies to minimize or avoid opioid use. Advise pregnant patients requiring long-term opioid use of the risk of NAS/NOWS and provide appropriate treatment for the neonate after delivery. NAS/NOWS is an expected and treatable condition following chronic opioid use during pregnancy and should not be the only reason to avoid treating pain with an opioid in pregnant patients (ACOG 2017; CDC [Dowell 2022]). Do not abruptly discontinue opioids during pregnancy; taper prior to discontinuation when appropriate, considering the risks to the pregnant patient and fetus if maternal withdrawal occurs (CDC [Dowell 2022]).

Monitoring Parameters

Acute pain (short-term): Pain relief, respiratory rate, mental status, blood pressure; stool output; signs of misuse, abuse, and substance use disorder.

Chronic pain (long-term therapy outside of end-of-life or palliative care, active cancer treatment, sickle cell disease, or medication-assisted treatment for opioid use disorder): Evaluate benefits/risks of opioid therapy within 1 to 4 weeks of treatment initiation and with dose increases. Reevaluate 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 with consideration for rechecking 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]).

Mechanism of Action

Binds to opiate receptors in the CNS, causing inhibition of ascending pain pathways, altering the perception of and response to pain; produces generalized CNS depression

Pharmacokinetics (Adult Data Unless Noted)

Onset of action: Pain relief: Immediate release: 10 to 15 minutes.

Peak effect: Immediate release: 0.5 to 1 hour.

Duration: Immediate release: 3 to 6 hours; Extended release: ≤12 hours.

Distribution: Vd: Children 2 to 10 years: 2.1 L/kg (range: 1.2 to 3.7 L/kg); Adults: 2.6 L/kg; distributed to skeletal muscle, liver, intestinal tract, lungs, spleen, and brain.

Protein binding: 38% to 45%.

Metabolism: Hepatically via CYP3A4 to noroxycodone (has weak analgesic activity), noroxymorphone, and alpha- and beta-noroxycodol. CYP2D6 mediated metabolism produces oxymorphone (has analgesic activity; low plasma concentrations [<15%]), alpha- and beta-oxymorphol.

Bioavailability: Extended release tablet, immediate release: 60% to 87%; Extended release capsule is not bioequivalent to extended release tablet; however AUC, is similar in a fed state.

Half-life:

Apparent: Immediate release: 3.2 to ~4 hours; Extended release tablet: 4.5 hours; Extended release capsule: 5.6 hours.

Elimination: Children 2 to 10 years: 1.8 hours (range: 1.2 to 3 hours); Adults: 3.7 hours.

Adults with CrCl <60 mL/minute: Half-life increases by 1 hour, but peak oxycodone concentrations increase by 50% and AUC increases by 60%.

Adults with mild to moderate hepatic impairment: Half-life increases by 2.3 hours, peak oxycodone concentrations increase by 50%, and AUC increases by 95%.

Time to peak, plasma: Immediate release: 1.2 to 1.9 hours; Extended release: 4 to 5 hours.

Excretion: Urine: (~10% as parent; ~65% as metabolites [noroxycodone (23%, active), oxymorphone (10%, active), noroxymorphone (14%, weakly active), reduced metabolites (≤18%)]) (Kinnunen 2019).

Pharmacokinetics: Additional Considerations (Adult Data Unless Noted)

Altered kidney function: Higher peak plasma oxycodone (50%), and noroxycodone (20%), higher AUC for oxycodone (60%), noroxycodone (50%), and oxymorphone (40%) in patients with CrCl <60 mL/minute. There is an increased half-life elimination for oxycodone elimination of only 1 hour.

Hepatic function impairment: Peak plasma oxycodone and noroxycodone concentrations 50% and 20% higher; AUC values are 95% and 65% higher, respectively, in mild to moderate hepatic impairment. Oxymorphone peak plasma concentration and AUC values are lower by 30% and 40%. The half-life elimination for oxycodone is increased by 2.3 hours.

Brand Names: International
International Brand Names by Country
For country code abbreviations (show table)

  • (AE) United Arab Emirates: Oxycontin | Oxynorm;
  • (AT) Austria: Oxycodon accord | Oxycodon hcl ratiopharm | Oxycontin | Oxygerolan | Oxynorm;
  • (AU) Australia: Genrx oxycodone mr | Gxp oxycodone | Novacodone | Oxycodone actavis | Oxycodone as | Oxycodone bnm | Oxycodone genpar | Oxycodone gppl | Oxycodone gx | Oxycodone medis | Oxycodone neo health | Oxycodone nhpl | Oxycodone sandoz | Oxycodone wt | Oxycodone zp | Oxycodor | Oxycontin | Oxylenus | Oxyndone | Oxynorm | Proladone | Xedone;
  • (BE) Belgium: Oxycodon sandoz | Oxycontin;
  • (BG) Bulgaria: Oxycodone | Oxycodone actavis | Oxylan;
  • (BR) Brazil: Cloridrato de oxicodona | Oxycontin | Oxygesic | Oxypynal;
  • (CH) Switzerland: Oxycodon actavis | Oxycodon sandoz | Oxycodon streuli | Oxycodone mepha retard | Oxynorm | Unox;
  • (CL) Chile: Nokotin | Oxycontin;
  • (CN) China: Oxycontin;
  • (CO) Colombia: Oxicodona clorhidrato | Oxycontin | Oxycontin orf;
  • (CZ) Czech Republic: Dolocodon | Oxycodon ratiopharm | Oxykodon | Oxykodon actavis | Oxykodon G. L. Pharma | Oxykodon stada;
  • (DE) Germany: Carenoxal | Oxycan uno | Oxycodon Hcl AbZ | Oxycodon HCl Actavis | Oxycodon Hcl AL | Oxycodon hcl awd | Oxycodon HCl beta | Oxycodon HCl CT | Oxycodon hcl dura | Oxycodon hcl hexal | Oxycodon HCL Hormosan | Oxycodon HCl Krugmann | Oxycodon HCl Krugmann akut | Oxycodon hcl mylan | Oxycodon hcl ratiopharm | Oxycodon hcl sandoz | Oxycodon hcl stada | Oxycodon hcl winthrop | Oxycodon hcl zentiva | Oxycodon painbreak | Oxycodone hcl acino | Oxycodone hcl aristo | Oxycodonhydrochlorid 1 A Pharma | Oxycodonhydrochlorid axcount | Oxycodonhydrochlorid Hennig | Oxycodonhydrochlorid Heumann | Oxycodonhydrochlorid puren | Oxyconoica | Oxygesic | Oxygesic akut | Oxynorm;
  • (EE) Estonia: Dolocodon | Oxycodone vitabalans;
  • (EG) Egypt: Oxynorm;
  • (ES) Spain: Oxicodona aristo | Oxicodona kern pharma | Oxicodona Sandoz | Oxynorm;
  • (FI) Finland: Oxycodon Hcl Accord | Oxycodone orion | Oxycodone Ratiopharm | Oxycodone sandoz | Oxycodone stada | Oxycodone vitabalans | Oxycorion depot | Oxynorm | Oxyratio;
  • (FR) France: Oxycodone accord LP | Oxycodone arrow | Oxycodone biogaran LP | Oxycodone Eg | Oxycodone mylan | Oxycodone sandoz | Oxycontin lp | Oxynorm;
  • (GB) United Kingdom: Carexil | Dolocodon | Ixyldone | Leveraxo | Longtec | Lynlor | Onexila XL | Oxeltra | Oxycodone | Oxycontin | Oxylan | Oxynorm | Oxypro | Proladone | Reltebon | Shortec | Zomestine;
  • (HK) Hong Kong: Oxycontin | Oxynorm;
  • (HR) Croatia: Oxycontin;
  • (HU) Hungary: Codoxy | Codoxy rapid | Oxycodone vitabalans | Oxycontin | Reltebon;
  • (ID) Indonesia: Oxyneo | Oxynorm;
  • (IE) Ireland: Dancex | Oxycodone | Oxycodone actavis | Oxycontin | Oxynorm | Reltebon;
  • (IL) Israel: Oxycontin;
  • (IT) Italy: Ossicodone aurobindo | Ossicodone Bruno Farmaceutici | Oxicodone accord | Oxicodone sandoz | Oxycontin;
  • (JP) Japan: Oxycodone | Oxycontin | Oxycontin dainippo;
  • (KR) Korea, Republic of: Ocodone cr | Oxycontin | Oxycontin cr | Procontin;
  • (LB) Lebanon: Oxycontin | Oxynorm;
  • (LT) Lithuania: Oxycodone;
  • (LU) Luxembourg: Oxycontin | Oxynorm;
  • (LV) Latvia: Oxycodone vitabalans | Oxycontin;
  • (MY) Malaysia: Oxycontin | Oxynorm;
  • (NL) Netherlands: Oxycodon | Oxycodon actavis | Oxycodon Hcl Accord | Oxycodon hcl gl | Oxycodon hcl mundipharma | Oxycodon hcl pch | Oxycodon hcl ratiopharm | Oxycodon hcl sandoz | Oxycodon HCl Teva | Oxycontin | Oxynorm;
  • (NO) Norway: Oxycodone actavis | Oxycodone orifarm | Oxycodone vitabalans | Oxycontin | Oxynorm | Reltebon;
  • (NZ) New Zealand: Oxycodone sandoz | Oxycontin | Oxydone Bnm | Oxynorm;
  • (PE) Peru: Nokotin | Oxirapid | Oxycontin | Oxydonna;
  • (PH) Philippines: Oxycontin;
  • (PL) Poland: Oxycodone vitabalans | Oxydolor Fast | Xancodal;
  • (PR) Puerto Rico: Oxycodone HCL | Oxycontin | Oxyir | Roxybond | Xtampza er;
  • (PT) Portugal: Olbete;
  • (PY) Paraguay: Sedalgina;
  • (QA) Qatar: Lynlor | OxyNorm;
  • (RO) Romania: Oxicodona Sandoz | Reltebon;
  • (SA) Saudi Arabia: Oxynorm;
  • (SE) Sweden: Enoxy Depot | Oxikodon acino | Oxikodon actavis | Oxikodon evolan | Oxycodone depot 1a farma | Oxycodone depot orion | Oxycodone g.l. | Oxycodone lannacher | Oxycodone orifarm | Oxycodone sandoz | Oxycodone teva | Oxycodone vitabalans | Oxyconta depot | Oxycontin | Oxynorm;
  • (SG) Singapore: Oxycontin;
  • (SI) Slovenia: Oksikodon vitabalans | Oxycontin;
  • (SK) Slovakia: Contiroxil | Dolocodon | Oxycodone pr actavis | Oxypro;
  • (TH) Thailand: Oxycontin;
  • (TR) Turkey: Oxopane;
  • (TW) Taiwan: Oxycontin | Oxynorm;
  • (UA) Ukraine: Dolonica;
  • (UG) Uganda: Oxynorm;
  • (ZA) South Africa: Oxycontin | Oxycorrell | Oxynorm
  1. 2023 American Geriatrics Society Beers Criteria Update Expert Panel. American Geriatrics Society 2023 updated AGS Beers Criteria for potentially inappropriate medication use in older adults. J Am Geriatr Soc. 2023;71(7):2052-2081. doi:10.1111/jgs.18372 [PubMed 37139824]
  2. Ahlfors CE. Benzyl alcohol, kernicterus, and unbound bilirubin. J Pediatr. 2001;139(2):317-319. [PubMed 11487763]
  3. Algera MH, Kamp J, van der Schrier R, et al. Opioid-induced respiratory depression in humans: a review of pharmacokinetic-pharmacodynamic modelling of reversal. Br J Anaesth. 2019;122(6):e168-e179. doi:10.1016/j.bja.2018.12.023 [PubMed 30915997]
  4. 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]
  5. American College of Obstetricians and Gynecologists’ (ACOG) Committee on Clinical Consensus–Obstetrics. Pharmacologic stepwise multimodal approach for postpartum pain management: ACOG clinical consensus no. 1. Obstet Gynecol. 2021;138(3):507-517. doi:10.1097/AOG.0000000000004517 [PubMed 34412076]
  6. American College of Obstetricians and Gynecologists’ (ACOG) Committee on Clinical Consensus–Obstetrics. Committee opinion no. 711: opioid use and opioid use disorder in pregnancy. Obstet Gynecol. 2017;130(2):e81-e94. doi:10.1097/AOG.0000000000002235 [PubMed 28742676]
  7. American Pain Society (APS). Pain assessment and management initiative (PAMI) dosing guide. Updated November 2016b.
  8. American Pain Society (APS). Principles of analgesic use. Seventh edition. 2016a.
  9. Anand KJ, Willson DF, Berger J, et al. Tolerance and withdrawal from prolonged opioid use in critically ill children. Pediatrics. 2010;125(5):e1208-e1225. doi:10.1542/peds.2009-0489 [PubMed 20403936]
  10. Anderson DT, Fritz KL, Muto JJ. Oxycontin: the concept of a "ghost pill" and the postmortem tissue distribution of oxycodone in 36 cases. J Anal Toxicol. 2002;26(7):448-459. [PubMed 12422999]
  11. APO-Oxycodone CR (oxycodone) [product monograph]. Toronto, Ontario, Canada: Apotex Inc; December 2021.
  12. Aronoff GR, Bennett WM, Berns JS, et al. Drug Prescribing in Renal Failure: Dosing Guidelines for Adults and Children, 5th ed. Philadelphia, PA: American College of Physicians; 2007.
  13. Association of Paediatric Anaesthetists of Great Britain and Ireland (APA). Good practice in postoperative and procedural pain management, 2nd edition. Paediatr Anaesth. 2012;22(Suppl 1):1-79.
  14. Azhar A, Kim YJ, Haider A, et al. Response to oral immediate-release opioids for breakthrough pain in patients with advanced cancer with adequately controlled background pain. Oncologist. 2019;24(1):125-131. doi:10.1634/theoncologist.2017-0583 [PubMed 30254187]
  15. Berde CB, Sethna NF. Analgesics for the treatment of pain in children. N Engl J Med. 2002;347(14):1094-1103. [PubMed 12362012]
  16. Bluthenthal RN, Simpson K, Ceasar RC, Zhao J, Wenger L, Kral AH. Opioid withdrawal symptoms, frequency, and pain characteristics as correlates of health risk among people who inject drugs. Drug Alcohol Depend. 2020;211:107932. doi:10.1016/j.drugalcdep.2020.107932 [PubMed 32199668]
  17. 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]
  18. Buys MJ. Use of opioids for postoperative pain control. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed March 21, 2023.
  19. Carr D. Management of acute pain in the patient chronically using opioids for non-cancer pain. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed October 11, 2021.
  20. Carvalho B, Sutton CD. Post-cesarean delivery analgesia. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed April 26, 2023.
  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. Centers for Disease Control and Prevention (CDC). Neonatal deaths associated with use of benzyl alcohol—United States. MMWR Morb Mortal Wkly Rep. 1982;31(22):290-291. http://www.cdc.gov/mmwr/preview/mmwrhtml/00001109.htm [PubMed 6810084]
  23. Center for Substance Abuse Treatment. Clinical Guidelines for the Use of Buprenorphine in the Treatment of Opioid Addiction. Rockville (MD): Substance Abuse and Mental Health Services Administration (US); 2004. [PubMed 22514846]
  24. Chang SH, Maney KM, Phillips JP, Langford RM, Mehta V. A comparison of the respiratory effects of oxycodone versus morphine: a randomised, double-blind, placebo-controlled investigation. Anaesthesia. 2010;65(10):1007-1012. doi:10.1111/j.1365-2044.2010.06498.x [PubMed 20712805]
  25. Cheung CW, Ching Wong SS, Qiu Q, Wang X. Oral oxycodone for acute postoperative pain: a review of clinical trials. Pain Physician. 2017;20(2S):SE33-SE52. [PubMed 28226340]
  26. Chou R, Gordon DB, de Leon-Casasola OA, et al. Management of postoperative pain: a clinical practice guideline from the American Pain Society, the American Society of Regional Anesthesia and Pain Medicine, and the American Society of Anesthesiologists' Committee on Regional Anesthesia, Executive Committee, and Administrative Council. J Pain. 2016;17(2):131-157. doi:10.1016/j.jpain.2015.12.008 [PubMed 26827847]
  27. Coté CJ, Lerman J, Anderson B, eds. A Practice of Anesthesia for Infants and Children. 6th ed. Elsevier; 2019.
  28. Crockett SD, Greer KB, Heidelbaugh JJ, et al. American Gastroenterological Association Institute guideline on the medical management of opioid-induced constipation. Gastroenterology. 2019;156(1):218-226. doi:10.1053/j.gastro.2018.07.016 [PubMed 30340754]
  29. Davison SN. Clinical pharmacology considerations in pain management in patients with advanced kidney failure. Clin J Am Soc Nephrol. 2019;14(6):917-931. doi:10.2215/CJN.05180418 [PubMed 30833302]
  30. De Aquino JP, Parida S, Sofuoglu M. The pharmacology of buprenorphine microinduction for opioid use disorder. Clin Drug Investig. 2021;41(5):425-436. doi:10.1007/s40261-021-01032-7 [PubMed 33818748]
  31. de Vries F, Bruin M, Lobatto DJ, et al. Opioids and their endocrine effects: a systematic review and meta-analysis. J Clin Endocrinol Metab. 2020;105(3):1020-1029. doi:10.1210/clinem/dgz022 [PubMed 31511863]
  32. Debono M, Chan S, Rolfe C, Jones TH. Tramadol-induced adrenal insufficiency. Eur J Clin Pharmacol. 2011; 67:865-867. doi:10.1007/s00228-011-0992-9 [PubMed 21243342]
  33. 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]
  34. Dowell D, Ragan KR, Jones CM, Baldwin GT, Chou R. CDC clinical practice guideline for prescribing opioids for pain - United States, 2022. MMWR Recomm Rep. 2022;71(3):1-95. doi:10.15585/mmwr.rr7103a1 [PubMed 36327391]
  35. Expert opinion. Senior Renal Editorial Team: Bruce Mueller, PharmD, FCCP, FASN, FNKF; Jason A. Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC; Michael Heung, MD, MS.
  36. Farmer AD, Drewes AM, Chiarioni G, et al. Pathophysiology and management of opioid-induced constipation: European expert consensus statement. United European Gastroenterol J. 2019;7(1):7-20. doi:10.1177/2050640618818305 [PubMed 30788113]
  37. Foral PA, Ineck JR, Nystrom KK. Oxycodone accumulation in a hemodialysis patient. South Med J. 2007;100(2):212-214. doi:10.1097/01.smj.0000242876.45747.0a [PubMed 17330696]
  38. Fox LM, Hoffman RS, Vlahov D, Manini AF. Risk factors for severe respiratory depression from prescription opioid overdose. Addiction. 2018;113(1):59-66. doi:10.1111/add.13925 [PubMed 28646524]
  39. Gadelha MR, Karavitaki N, Fudin J, Bettinger JJ, Raff H, Ben-Shlomo A. Opioids and pituitary function: expert opinion. Pituitary. 2022;25(1):52-63. doi:10.1007/s11102-021-01202-y [PubMed 35066756]
  40. Gallagher RM, Welz-Bosna M, Gammaitoni A. Assessment of dosing frequency of sustained-release opioid preparations in patients with chronic nonmalignant pain. Pain Med. 2007;8(1):71-74. [PubMed 17244106]
  41. Godwin B, Frank C, Molnar F, Dyks D, Akter R. Identification and management of opioid-induced neurotoxicity in older adults. Can Fam Physician. 2022;68(4):269-270. doi:10.46747/cfp.6804269 [PubMed 35418392]
  42. Gupta K, Nagappa M, Prasad A, et al. Risk factors for opioid-induced respiratory depression in surgical patients: a systematic review and meta-analyses. BMJ Open. 2018;8(12):e024086. doi:10.1136/bmjopen-2018-024086 [PubMed 30552274]
  43. Hellinga M, Algera MH, Olofsen E, et al. Oral oxycodone-induced respiratory depression during normocapnia and hypercapnia: A pharmacokinetic-pharmacodynamic modeling study. Clin Pharmacol Ther. 2023;113(5):1080-1088. doi:10.1002/cpt.2863 [PubMed 36744649]
  44. 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]
  45. Hjalte F, Berggren AC, Bergendahl H, Hjortsberg C. The direct and indirect costs of opioid-induced constipation. J Pain Symptom Manage. 2010;40(5):696-703. doi:10.1016/j.jpainsymman.2010.02.019 [PubMed 20727708]
  46. Hudak ML, Tan RC, Committee on Drugs, et al, "Neonatal Drug Withdrawal," Pediatrics, 2012, 129(2):e540-60. [PubMed 22291123]
  47. "Inactive" ingredients in pharmaceutical products: update (subject review). American Academy of Pediatrics (AAP) Committee on Drugs. Pediatrics. 1997;99(2):268-278. [PubMed 9024461]
  48. Jilani SM, Jones HE, Grossman M, et al. Standardizing the clinical definition of opioid withdrawal in the neonate. J Pediatr. 2022;243:33-39.e1. doi:10.1016/j.jpeds.2021.12.021 [PubMed 34942181]
  49. Khanna AK, Bergese SD, Jungquist CR, et al. Prediction of opioid-induced respiratory depression on inpatient wards using continuous capnography and oximetry: An international prospective, observational trial. Anesth Analg. 2020;131(4):1012-1024. doi:10.1213/ANE.0000000000004788 [PubMed 32925318]
  50. Kinnunen M, Piirainen P, Kokki H, Lammi P, Kokki M. Updated clinical pharmacokinetics and pharmacodynamics of oxycodone. Clin Pharmacokinet. 2019;58(6):705-725. doi:10.1007/s40262-018-00731-3 [PubMed 30652261]
  51. Kirvela M, Lindgren L, Seppala T, Olkkola KT. The pharmacokinetics of oxycodone in uremic patients undergoing renal transplantation. J Clin Anesth. 1996;8(1):13-18. doi:10.1016/0952-8180(95)00092-5 [PubMed 8695073]
  52. Kokki M, Franco MG, Raatikainen K, et al. Intravenous oxycodone for pain relief in the first stage of labour--maternal pharmacokinetics and neonatal exposure. Basic Clin Pharmacol Toxicol. 2012;111(3):182-188. [PubMed 22448718]
  53. 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]
  54. Kosten TR, Baxter LE. Review article: Effective management of opioid withdrawal symptoms: A gateway to opioid dependence treatment. Am J Addict. 2019;28(2):55-62. doi:10.1111/ajad.12862 [PubMed 30701615]
  55. Lam J, Kelly L, Ciszkowski C, et al. Central nervous system depression of neonates breastfed by mothers receiving oxycodone for postpartum analgesia. J Pediatr. 2012;160(1):33-37. [PubMed 21880331]
  56. Leuppi-Taegtmeyer A, Duthaler U, Hammann F, et al. Pharmacokinetics of oxycodone/naloxone and its metabolites in patients with end-stage renal disease during and between haemodialysis sessions. Nephrol Dial Transplant. 2019;34(4):692-702. doi:10.1093/ndt/gfy285 [PubMed 30189012]
  57. Lozano-López MT, Gamonal-Limcaoco S, Casado-Espada N, et al. Psychosis after buprenorphine, heroin, methadone, morphine, oxycodone, and tramadol withdrawal: A systematic review. Eur Rev Med Pharmacol Sci. 2021;25(13):4554-4562. doi:10.26355/eurrev_202107_26248 [PubMed 34286498]
  58. Maffei MV, Laehn S, Bianchini M, Kim A. Risk factors associated with opioid/benzodiazepine iatrogenic withdrawal syndrome in COVID-19 acute respiratory distress syndrome. J Pharm Pract. 2022;8971900221116178. doi:10.1177/08971900221116178 [PubMed 35930693]
  59. Marcus DA, Glick RM. Sustained-release oxycodone dosing survey of chronic pain patients. Clin J Pain. 2004;20(5):363-366. [PubMed 15322444]
  60. 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]
  61. McMillan SC. Assessing and managing opiate-induced constipation in adults with cancer. Cancer Control. 2004;11(3 Suppl):3-9. doi:10.1177/10732748040110S302 [PubMed 15153834]
  62. Meek JY, Noble L; Section on Breastfeeding. Policy statement: Breastfeeding and the use of human milk. Pediatrics. 2022;150(1):e2022057988. doi:10.1542/peds.2022-057988 [PubMed 35921640]
  63. Mercadante S. Pathophysiology and treatment of opioid-related myoclonus in cancer patients. Pain. 1998;74(1):5-9. [PubMed 9514554]
  64. Montandon G. The pathophysiology of opioid-induced respiratory depression. Handb Clin Neurol. 2022;188:339-355. doi:10.1016/B978-0-323-91534-2.00003-5 [PubMed 35965031]
  65. Müller-Lissner S, Bassotti G, Coffin B, et al. Opioid-induced constipation and bowel dysfunction: A clinical guideline. Pain Med. 2017;18(10):1837-1863. doi:10.1093/pm/pnw255 [PubMed 28034973]
  66. Nicholson B, Ross E, Sasaki J, Weil A. Randomized trial comparing polymer-coated extended-release morphine sulfate to controlled-release oxycodone HCl in moderate to severe nonmalignant pain. Curr Med Res Opin. 2006;22(8):1503-1514. [PubMed 16870075]
  67. Oxaydo (oxycodone) [prescribing information]. Wayne, PA: Zyla Life Sciences US Inc; March 2021.
  68. OxyContin (oxycodone) extended-release tablets [prescribing information]. Stamford, CT: Purdue Pharma LP; October 2021.
  69. Oxycodone hydrochloride capsules [prescribing information]. Allentown, PA: Genus Lifesciences Inc; July 2021.
  70. Oxycodone hydrochloride 1 mg/mL oral solution [prescribing information]. Allentown, PA: Genus Lifesciences Inc; July 2021.
  71. Oxycodone hydrochloride oral solution [prescribing information]. Berkeley Heights, NJ: Hikma Pharmaceuticals USA Inc; July 2021.
  72. Oxycodone hydrochloride oral solution [prescribing information]. Largo, FL: VistaPharm; March 2021.
  73. Oxycodone hydrochloride tablet [prescribing information]. Coventry, RI: Rhodes Pharmaceuticals LP; March 2021.
  74. Oxy IR (oxycodone) [product monograph]. Toronto, Ontario, Canada: Purdue Pharma; October 2022.
  75. Oxyneo (oxycodone) controlled release tablets [product monograph]. Toronto, Ontario, Canada: Purdue Pharma; July 2022.
  76. Paice JA, Ferrell B. The management of cancer pain. CA Cancer J Clin. 2011;61(3):157-182. doi:10.3322/caac.20112 [PubMed 21543825]
  77. Pain Assessment and Management Initiative. Pain management & dosing guide. https://pami.emergency.med.jax.ufl.edu/wordpress/files/2020/09/PAMI-Dosing-Guide-Nov.2.2020.pdf. Updated November 2020. Accessed October 12, 2021.
  78. Pappagallo M. Incidence, prevalence, and management of opioid bowel dysfunction. Am J Surg. 2001;182(suppl 5A):11S-18S. doi:10.1016/s0002-9610(01)00782-6 [PubMed 11755892]
  79. Patrick SW, Barfield WD, Poindexter BB; Committee on Fetus and Newborn; Committee on Substance Use and Prevention. Neonatal opioid withdrawal syndrome. Pediatrics. 2020;146(5):e2020029074. doi:10.1542/peds.2020-029074 [PubMed 33106341]
  80. Pattinson KT. Opioids and the control of respiration. Br J Anaesth. 2008;100(6):747-758. doi:10.1093/bja/aen094 [PubMed 18456641]
  81. Pattullo GG. Clinical implications of opioid-induced ventilatory impairment. Anaesth Intensive Care. 2022;50(1-2):52-67. doi:10.1177/0310057X211070292 [PubMed 35189729]
  82. Pino CA, Wakerman 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.
  83. Portenoy RK, Mehta Z, Ahmed E. Cancer pain management with opioids: optimizing analgesia. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed October 12, 2021.
  84. Poulsen JL, Brock C, Olesen AE, Nilsson M, Drewes AM. Clinical potential of naloxegol in the management of opioid-induced bowel dysfunction. Clin Exp Gastroenterol. 2014;7:345-358. doi:10.2147/CEG.S52097 [PubMed 25278772]
  85. Rakoski M, Goyal P, Spencer-Safier M, Weissman J, Mohr G, Volk M. Pain management in patients with cirrhosis. Clin Liver Dis (Hoboken). 2018;11(6):135-140. doi:10.1002/cld.711 [PubMed 30992804]
  86. 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]
  87. Refer to manufacturer's labeling.
  88. 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.
  89. Rossi M, Casale G, Badiali D, et al. Opioid-induced bowel dysfunction: suggestions from a multidisciplinary expert board. Support Care Cancer. 2019;27(11):4083-4090. doi:10.1007/s00520-019-04688-2 [PubMed 30778756]
  90. Roxicodone (oxycodone) [prescribing information]. Webster Groves, MO: SpecGx LLC; January 2023.
  91. Roxybond (oxycodone) [prescribing information]. Princeton, NJ: Protega Pharmaceuticals LLC; February 2022.
  92. Sachs HC, Committee On Drugs. The transfer of drugs and therapeutics into human breast milk: an update on selected topics. Pediatrics. 2013;132(3):e796-809. [PubMed 23979084]
  93. Seaton S, Reeves M, McLean S. Oxycodone as a component of multimodal analgesia for lactating mothers after Caesarean section: relationships between maternal plasma, breast milk and neonatal plasma levels. Aust N Z J Obstet Gynaecol. 2007;47(3):181-185. [PubMed 17550483]
  94. Smith HS. Opioid metabolism. Mayo Clin Proc. 2009;84(7):613-624. doi:10.1016/S0025-6196(11)60750-7 [PubMed 19567715]
  95. Smith HAB, Besunder JB, Betters KA, et al. 2022 Society of Critical Care Medicine clinical practice guidelines on prevention and management of pain, agitation, neuromuscular blockade, and delirium in critically ill pediatric patients with consideration of the ICU environment and early mobility. Pediatr Crit Care Med. 2022;23(2):e74-e110. doi:10.1097/PCC.0000000000002873 [PubMed 35119438]
  96. Spadaro A, Faude S, Perrone J, et al. Precipitated opioid withdrawal after buprenorphine administration in patients presenting to the emergency department: A case series. J Am Coll Emerg Physicians Open. 2023;4(1):e12880. doi:10.1002/emp2.12880 [PubMed 36704210]
  97. Spadaro A, Long B, Koyfman A, Perrone J. Buprenorphine precipitated opioid withdrawal: Prevention and management in the ED setting. Am J Emerg Med. 2022;58:22-26. doi:10.1016/j.ajem.2022.05.013 [PubMed 35623179]
  98. Sulton-Villavasso C, Austin CA, Patra KP, et al. Index of suspicion. Case 1: Infant who has respiratory distress. Case 2: Abnormal behavior, seizures, and altered sensorium in a 7-year-old boy. Case 3: Fever and dysphagia in a 4-year-old girl. Pediatr Rev. 2012;33(6):279-284. [PubMed 22659261]
  99. Supeudol (oxycodone) [product monograph]. Boucherville, Quebec, Canada: Sandoz Canada Inc; May 2023.
  100. Tamayo AC, Diaz-Zuluaga PA. Management of opioid-induced bowel dysfunction in cancer patients. Support Care Cancer. 2004;12(9):613-618. doi:10.1007/s00520-004-0649-7 [PubMed 15221581]
  101. Tawfic QA, Bellingham G. Postoperative pain management in patients with chronic kidney disease. J Anaesthesiol Clin Pharmacol. 2015;31(1):6-13. doi:10.4103/0970-9185.150518 [PubMed 25788766]
  102. Tran BW, Kohan LR, Vorenkamp KE. Postoperative oxycodone toxicity in a patient with chronic pain and end-stage renal disease. A A Case Rep. 2015;4(4):44-46. doi:10.1213/XAA.0000000000000115 [PubMed 25689360]
  103. Tuteja AK, Biskupiak J, Stoddard GJ, Lipman AG. Opioid-induced bowel disorders and narcotic bowel syndrome in patients with chronic non-cancer pain. Neurogastroenterol Motil. 2010;22(4):424-e96. doi:10.1111/j.1365-2982.2009.01458.x [PubMed 20100280]
  104. US Department of Health and Human Services (HHS). HHS guide for clinicians on the appropriate dosage reduction or discontinuation of long-term opioid analgesics. https://www.hhs.gov/opioids/sites/default/files/2019-10/Dosage_Reduction_Discontinuation.pdf. Updated October 2019. Accessed October 12, 2021.
  105. US Food and Drug Administration (FDA). FDA drug safety communication: FDA updates prescribing information for all opioid pain medicines to provide additional guidance for safe use. https://www.fda.gov/drugs/drug-safety-and-availability/fda-updates-prescribing-information-all-opioid-pain-medicines-provide-additional-guidance-safe-use. Published April 13, 2023. Accessed April 17, 2023.
  106. van der Schrier R, Jonkman K, van Velzen M, et al. An experimental study comparing the respiratory effects of tapentadol and oxycodone in healthy volunteers. Br J Anaesth. 2017;119(6):1169-1177. doi:10.1093/bja/aex295 [PubMed 29029015]
  107. Wang PP, Huang E, Feng X, et al. Opioid-associated iatrogenic withdrawal in critically ill adult patients: a multicenter prospective observational study. Ann Intensive Care. 2017;7(1):88. doi:10.1186/s13613-017-0310-5 [PubMed 28866754]
  108. Weinberg DS, Inturrisi CE, Reidenberg B, et al. Sublingual absorption of selected opioid analgesics. Clin Pharmacol Ther. 1988;44(3):335-342. doi:10.1038/clpt.1988.159 [PubMed 2458208]
  109. World Health Organization (WHO). Breastfeeding and maternal medication, recommendations for drugs in the eleventh WHO model list of essential drugs. 2002. Available at https://apps.who.int/iris/handle/10665/62435
  110. Xtampza ER (oxycodone) [prescribing information]. Cincinnati, OH: Patheon Pharmaceuticals; March 2021.
Topic 12666 Version 717.0

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟