Expired activity
Please go to the PowerPak homepage and select a course.

Building Pharmacist Skills in Opioid Analgesic Therapy: Opioid Analgesics: An Overview of Properties and Formulations

Introduction

Morphine, the prototypical opioid drug, was first isolated from the opium poppy in 1803.1 Since then, a number of additional opioid drugs have been isolated or synthesized. Opioids are commonly used to treat pain, but different opioid analgesics have different properties and indications.1,2 There are also a number of different opioid formulations available, including formulations that combine opioid and nonopioid analgesics. It is essential for pharmacists to understand the differences between these drugs and formulations, so that they can recommend appropriate therapy for patients.

Mechanisms of Action for Opioid Analgesics

Opioid analgesics are so named because they act at opioid receptors, which are found primarily on neurons in the central nervous system and gastrointestinal tract.3 These receptors can also be found on neurons throughout the periphery and on other cell types, including macrophages and astrocytes.2 Opioid receptors are G-protein coupled receptors that, when activated, produce several changes in neuronal cells.2,4 The activation of opioid receptors leads to a decreased opening of voltage-gated calcium channels, which in turn reduces the release of neurotransmitters from presynaptic terminals. Opioid receptor activation also stimulates potassium currents, which hyperpolarizes the postsynaptic neuron and thereby suppresses neuronal activity.

The pain-relieving effects of opioid analgesics are primarily attributed to their binding at opioid receptors in the brain and spinal cord.2 In the brain, an area known as the mesencephalic periaqueductal gray (PAG) contains opioid-sensitive pathways. Activation of opioid receptors in this area blocks the release of gamma-aminobutyric acid (GABA) from tonically active systems that regulate activity in projections to the nucleus raphe magnus (NRM) in the medulla.2,4 Increased activity in these projections causes an increase in the release of serotonin and norepinephrine from descending inhibitory neurons in the dorsal horn of the spine, resulting in reduced dorsal horn excitability. Opioids additionally act on the nucleus reticularis paragigantocellularis (NRPG) in the medulla, which similarly stimulates increased inhibitory outflow from the NRM to the spinal dorsal horn.4 Opioids can also act directly at the spinal level to inhibit the firing of pain transmission neurons in the dorsal horn and inhibit excitatory neurotransmitter release from primary afferent neurons.1

There are 3 major classes of opioid receptors: mu opioid receptors (MOR), delta opioid receptors (DOR), and kappa opioid receptors (KOR).2 Mu opioid receptors are the primary receptors responsible for the analgesic and euphoric effects of opioids.5 These receptors are also thought to be responsible for some of the most well-known side effects of opioid analgesics (constipation and respiratory depression). Activation of DOR provides limited analgesia in acute pain, but may play a larger role in the relief of chronic pain. DOR agonism can also have antidepressant and proconvulsant effects. Agonists at KOR can cause either analgesic or proalgesic effects, depending on the agonist. Activation of KOR can also lead to dysphoria, depression, and anticonvulsant effects. Most opioid analgesics used in clinical practice exert their therapeutic effects through MOR agonism, but they typically have some degree of activity at the other opioid receptors as well.2,5 Different opioid analgesic agents have different degrees of activity at various receptors (Table 1).1,2

Some opioids are termed “partial agonists” because they bind specifically to opioid receptors but have limited intrinsic activity at those receptors.2 These drugs include buprenorphine, nalbuphine, pentazocine, and butorphanol. Buprenorphine is a partial agonist at MOR and an antagonist at DOR and KOR.1,2 Because of its mixed activity, it may precipitate withdrawal in patients who are physically dependent on full agonist opioid analgesics. Nalbuphine is an antagonist at MOR, but exerts an analgesic effect through strong agonist activity at KOR. Pentazocine also exhibits agonist activity at KOR, with partial agonist or weak antagonist activity at MOR. Butorphanol is primarily a KOR agonist, but it may also act as an MOR partial agonist or antagonist.1

Some opioid agents that primarily act through MOR agonism have additional modes of action as well. Methadone can block N-methyl-D-aspartate (NMDA) receptors and monoaminergic reuptake transporters in addition to acting as a MOR agonist.1 This may allow it to be more effective for pain that is difficult to treat, such as cancer-related pain or neuropathic pain, and pain that does not respond adequately to morphine. Levorphanol also has some NMDA antagonist properties and inhibits reuptake of serotonin and norepinephrine.

Tramadol and tapentadol have activity at opioid receptors, but it is thought that they produce their primary analgesic effects via other mechanisms.1 Tramadol is a centrally acting analgesic with a complex mechanism of action. It acts as a weak MOR agonist, but a significant portion of its analgesic effect is derived from its ability to inhibit serotonin and norepinephrine reuptake. Tapentadol has stronger activity at MOR than tramadol, but it also significantly inhibits norepinephrine reuptake.

Some opioid agents are more potent than others, meaning that they bind to opioid receptors more strongly.2,6 In general, fentanyl, sufentanil, and buprenorphine are considered high potency opioids, while methadone, oxycodone, morphine, and hydromorphone are considered medium potency and codeine, hydrocodone, tramadol, and tapentadol are considered low potency.6 Opioids that are more potent require lower doses to produce a similar analgesic effect (Table 2).7 It is worth noting that “equianalgesic” doses from a potency perspective are approximate and may not be directly equivalent for therapeutic purposes: however, a full discussion of opioid conversion in practice is beyond the scope of this module.

Table 1. Opioid Receptor Actions of Clinically Used Opioid Analgesics.1,2
Opioid Analgesic MOR Activity DOR Activity KOR Activity Other Actions
Buprenorphine PA - -  
Butorphanol PA   +++  
Codeine +      
Dihydrocodeine +      
Fentanyl +++      
Hydrocodone +      
Hydromorphone +++   +  
Levorphanol +++     Block NMDA receptors

Block serotonin/NE reuptake
Meperidine +++      
Methadone +++     Block NMDA receptors

Block monoaminergic reuptake transporters
Morphine +++   +  
Nalbuphine -   ++  
Oxycodone ++      
Oxymorphone +++      
Pentazocine PA   +  
Sufentanil +++ + +  
Tapentadol ++     Block NE reuptake
Tramadol +     Block serotonin/NE reuptake
Abbreviations: DOR, delta opioid receptor; KOR, kappa opioid receptor; MOR, mu opioid receptor; NE, norepinephrine; NMDA, N-methyl-D-aspartate; PA, partial agonist.
Table 2. Approximate Equianalgesic Opioid Dosing.7
  Equianalgesic doses (mg)a
Opioid Analgesic Oral Parenteral
Morphine 25 10
Codeine 200 100
Fentanyl NA 0.15
Hydrocodone 25 NA
Hydromorphone 5 2
Meperidine 300 100
Oxycodone 20 10
Oxymorphone 10 1
Tapentadol 100 NA
Tramadol 120 100
a Presented to demonstrate relative potencies of various opioid drugs; “equianalgesic” doses are approximate and may not be therapeutically equivalent in practice.

Abbreviations: NA, not applicable.

Short-Acting Opioid Analgesics: Immediate-Release Formulations and Combination Products

A number of opioid analgesics are available in immediate-release formulations.8 These may be single product formulations, or combination formulations that include one or more nonopioid analgesics in addition to the opioid agent. Table 3 provides an overview of the available short-acting products, including indications and dosing recommendations. Short-acting formulations are typically used for acute pain and pain that is not consistently present (for example, pain due to injury or surgery, or breakthrough pain in patients who are taking long-acting opioids for chronic cancer pain).9,10 Short-acting formulations are also recommended when first initiating opioid therapy for chronic pain; once an effective dose of the short-acting opioid is found, the patient can be converted to a long-acting formulation.9-11

Many short-acting agents come in multiple formulations.12 A route or formulation may be selected based on patient characteristics and the desired onset of action. For example, if a patient is experiencing acute, severe pain (ie, cancer breakthrough pain, postoperative pain, sickle cell pain crisis), immediate pain relief is required. The onset of action for oral pain medications is usually around 45 minutes, and the peak analgesic effects are not achieved until 1 to 2 hours after administration. Therefore, an alternate route of administration (such as the intravenous route or transmucosal route) would be more appropriate for patients who require immediate pain relief. Some patients may not be able to swallow tablets, and some patients may not be able to take medication orally at all. These patients will also require alternative formulations (eg, oral solutions, rectal suppositories, parenteral formulations).

Single Agent Immediate-Release Opioid Analgesics

Morphine can be given via oral, parenteral, and rectal routes.8,13 Onset of action varies by route. When administered orally, morphine has a bioavailability of approximately 20% to 40%.8 Onset of analgesia occurs after about 30 minutes, and peak analgesic effect is reached in about an hour.8,13 Intravenous injection results in a faster onset (5 to 10 minutes), with peak analgesia within 20 minutes. Intramuscular administration achieves peak analgesia in 30 to 60 minutes, while subcutaneous administration achieves peak analgesia in 50 to 90 minutes. Rectal morphine reaches its peak effect within 20 to 60 minutes. Morphine’s effects last for about 3 to 6 hours when given via most routes. However, the effects of rectal morphine may last for up to 7 hours, and epidural or intrathecal morphine can produce analgesia for up to 24 hours.13

Morphine has 2 major metabolites, morphine-3-glucuronide (M3G) and morphine-6-glucuronide (M6G).12 These metabolites are active; M6G contributes to analgesia, while M3G can cause undesirable central nervous system effects, such as seizures.2,8,12 Both metabolites are renally cleared, so they may accumulate in patients with renal impairment.12 It is generally recommended to avoid morphine in renally impaired patients.

Codeine itself has low affinity for opioid receptors and therefore lacks a strong analgesic effect.2,8 However, approximately 10% of the administered codeine dose is metabolized to morphine by CYP2D6 in the liver. It is thought that this conversion is responsible for the bulk of codeine’s analgesic effects. Polymorphisms in CYP2D6 can cause variations in response to codeine.2 Some patients may not respond to codeine at all, while others (termed “ultrarapid metabolizers”) may have increased codeine sensitivity. The onset of action for codeine is 30 to 60 minutes.8,13 Peak effects are achieved at 60 to 90 minutes post-dose, and effects last for 4 to 6 hours.

Hydromorphone is a semisynthetic derivative of morphine.2 It has a bioavailability of 60% when given orally, and onset of analgesia occurs within 15 to 30 minutes.8,13 Peak effect is achieved at 30 to 60 minutes. Intravenous administration allows for a 5-minute onset of action, with peak effect achieved within 10 to 20 minutes.13 The duration of action is approximately 3 to 4 hours for all immediate-release formulations, but the suppository may provide a longer duration of effect. Oxymorphone is a potent semisynthetic opioid with an oral bioavailability of 10%.8 Bioavailability may be significantly increased in moderate to severe hepatic impairment.

Oxycodone is a semisynthetic opioid commonly used for moderate-to-severe pain.8,12 Although it is metabolized to oxymorphone in the liver, its analgesic action is primarily attributed to the parent compound.8 Oxycodone’s first-pass metabolism is less extensive than that of many other opioids, so oral bioavailability is 60% to 87%. Onset of action occurs about 15 minutes after administration, and maximum effect occurs about 1 to 2 hours post-dose.8,13 Effects last for 3 to 6 hours.13

Fentanyl has several unique applications in pain management. It is highly lipid-soluble, which allows it to pass through the blood-brain barrier quickly.2 It has a quick onset of action and a rapid offset of action when given in small boluses (with high doses or prolonged infusions, fentanyl may begin to accumulate, resulting in a longer duration of action). Fentanyl can be administered transmucosally or intranasally for the treatment of breakthrough cancer pain.8,13 Recommendations for dosing and titration vary by product, and products cannot be directly interchanged. When administered transmucosally, it typically takes effect within 5 to 15 minutes and reaches peak effect in about 20 to 90 minutes, depending on the specific formulation used.13 The intranasal spray takes effect within 5 to 10 minutes and reaches peak effect in 15 to 21 minutes. Fentanyl is also available as a solution for injection, which is typically used as an adjunct to anesthesia or for pain associated with surgery or critical illness. Peak concentrations are achieved in minutes with intravenous administration, and effects last for 30 to 60 minutes.8

Intravenous sufentanil is primarily used in the context of anesthesia, but a sublingual formulation was approved for acute severe pain in 2018.13 This formulation is 53% bioavailable, and it takes effect within 30 minutes, reaching peak effect in 1 hour.8,13 Effects usually last about 3 hours.13 Sufentanil is several times more potent than fentanyl and should only be used under direct medical supervision.8

Meperidine can be used for acute pain, but it is not recommended for chronic use, because its active metabolite normeperidine can accumulate and increase the risk of seizures, particularly in renally impaired or elderly patients.12,13 Although oral meperidine is available, it is typically not recommended for use, because extensive first-pass metabolism can lead to high levels of normeperidine.13 Parenteral meperidine takes effect in 5 to 15 minutes, with peak effects in 5 to 7 minutes (intravenous) or 1 hour (intramuscular and subcutaneous). Effects last 2 to 4 hours.

Tramadol undergoes first-pass metabolism and has a bioavailability of 68%; however, bioavailability approaches 100% after multiple doses due to saturable first-pass metabolism.8 One of tramadol’s metabolites is active and contributes significantly to its analgesic effect. Onset of analgesic action occurs within 1 hour, with peak effects achieved in 2 to 3 hours. Analgesia lasts for 6 hours.

Butorphanol is available as an intranasal spray as well as an injectable formulation.8 The nasal spray can produce analgesia within 15 minutes, and it has a longer duration of action than intravenous or intramuscular administration (4-5 hours vs 2-4 hours). Peak effects are achieved in 1 to 2 hours with intranasal administration.13 Tapentadol has an oral bioavailability of about 32%, and absorption may increase with food.8 Peak plasma concentrations occur at 1.25 hours, and its half-life is approximately 4 hours.13

Nalbuphine must be administered parenterally, because it is metabolized in the gastrointestinal mucosa and it undergoes extensive first-pass metabolism.8 When administered intravenously, it has a rapid onset of action (2-3 minutes), with peak effects in 30 minutes. The onset of action with subcutaneous or intramuscular administration is around 15 minutes, and the duration of action for all parenteral routes is 3 to 6 hours.8,13

Combination Immediate-Release Opioid Analgesic Formulations

Combination therapy with medications from different therapeutic classes is commonly employed as a pain management strategy.12,14 Combining opioid analgesics with other pain-relieving drugs can help maintain or increase analgesic effectiveness while reducing the risk of opioid-induced adverse effects and allowing for lower doses of the opioid. Opioid analgesics are often coformulated with nonopioid analgesics or other drugs that may complement their pain-relieving actions.8

Coformulations with acetaminophen, ibuprofen, aspirin, carisoprodol, butalbital, and caffeine are available (Table 3).8 Acetaminophen, ibuprofen, and aspirin are nonopioid analgesics. Carisoprodol is a centrally acting muscle relaxant. Butalbital is a barbiturate: it enhances GABA activity in the thalamus, producing sedative and relaxant effects. Caffeine enhances the effects of analgesics and acts as a central nervous system stimulant.8,13 Of note, in 2011, the US Food and Drug Administration (FDA) limited the amount of acetaminophen that could be included in a single dosage form (ie, in 1 tablet or capsule) to 325 mg due to the risk of liver injury with acetaminophen overdose.15 When using a coformulated product that contains acetaminophen, it is important to ensure that the patient does not receive more than 4 g acetaminophen per day. Some combination products are only indicated for the short-term treatment of pain.8

Hydrocodone is a short-acting opioid, but it is not available as a single agent immediate-release product.8 It is only available in combination with acetaminophen or ibuprofen. Benzhydrocodone is a prodrug of hydrocodone that is only available as a coformulated tablet with acetaminophen. Benzhydrocodone is metabolized to hydrocodone by enzymes in the gastrointestinal tract, so systemic exposure to benzhydrocodone is minimal.

In some cases, short-acting opioid drugs may be coformulated with opioid antagonists to prevent abuse.14 Pentazocine is available in combination with naloxone, an opioid receptor antagonist.8 When administered orally, naloxone is poorly absorbed and has no significant activity; thus, if pentazocine/naloxone is taken orally, naloxone will not interfere with the action of pentazocine. However, if the tablet is crushed and injected intravenously, naloxone will counteract the effects of pentazocine and prevent the user from experiencing a “high.”8,14 This combination formulation is generally used as a second-line option for moderate-to-severe pain.8,12 When given orally, pentazocine typically takes effect within 15 to 30 minutes, and analgesic effects last for 3 hours or more.8

Table 3. Short-Acting Opioid Analgesic Formulations.8,12,13  
Generic Name and Formulations/Strengths FDA-Approved Indication(s) Typical Adult Dosing for Paina Notes
Single Agent Products

Butorphanol

 

Nasal spray: 10 mg/mL

 

Solution for injection: 1 mg/mL, 2 mg/mL

 

CIV

Nasal spray: Management of pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

Solution for injection: Management of pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate; management of labor pain; preoperative medication; adjunct to balanced anesthesia

Nasal spray:

1 mg (1 spray) into 1 nostril; if pain relief is not achieved in 60-90 min, another 1 mg spray can be administered in the other nostril; this 2-dose sequence can be repeated 3-4 h after the last dose PRN; a 2 mg dose (1 spray in each nostril) can be given initially, if the patient can remain recumbent; if a 2 mg dose is initially given, another dose should not be given for 3-4 h

 

IV:

For pain, 0.5-2 mg IV every 3-4 h PRN; for labor pain, 1 to 2 mg IV, may repeat in 4 h (alternative analgesia should be used if delivery is expected to occur within 4 h); when used as a supplement to anesthesia, 2 mg IV shortly before induction and/or incremental dose of 0.5-1 mg IV, up to 0.06 mg/kg (usual total dose, 4-12.5 mg)

 

IM:

For pain, 1-4 mg IM every 3-4 h PRN; for labor pain, 1-2 mg IM, may repeat in 4 h (alternative analgesia should be used if delivery is expected to occur within 4 h); for anesthetic premedication, 2 mg IM 60-90 min before surgery

 

 

In elderly patients and patients with renal or hepatic dysfunction, the nasal spray should be dosed as follows: 1 mg initially; a second dose may be administered after 90-120 min if needed; dosing sequence should generally not be repeated for at least 6 h. IV dosing should start at 0.5 mg, and IM dosing should start at 1 mg for these patients, with doses generally given at least 6 h apart

Codeine

 

Tablet: 15 mg, 30 mg, 60 mg

 

CII

Mild-to-moderate pain, where treatment with an opioid is appropriate

15-60 mg PO every 4 h PRN

 

Max: 60 mg/dose, 360 mg/24 h

Patients with prior opioid exposure may require higher initial doses

 

Use caution in severe renal impairment

 

Take with full glass of water; may take with food to decrease stomach irritation

Fentanyl

 

Buccal tablet: 100 mcg, 200 mcg, 400 mcg, 600 mcg, 800 mcg

 

Lozenge: 200 mcg, 400 mcg, 600 mcg, 800 mcg, 1200 mcg, 1600 mcg

 

Nasal spray: 100 mcg/actuation, 300 mcg/actuation, 400 mcg/actuation

 

Sublingual tablet: 100 mcg, 200 mcg, 300 mcg, 400 mcg, 600 mcg, 800 mcg

 

Sublingual spray: 100 mcg/spray, 200 mcg/spray, 400 mcg/spray, 600 mcg/spray, 800 mcg/spray, 1200 mcg/spray, 1600 mcg/spray

 

Solution for injection: 50 mcg/mL

 

CII

Buccal tablet, lozenge, nasal spray, sublingual tablet and spray: Management of breakthrough cancer pain in opioid-tolerant patients who are already receiving around-the-clock opioid therapy

 

Solution for injection: adjunct to anesthesia; use for premedication, analgesia during anesthesia, and in the immediate postoperative period

Buccal tablet:

100 mcg initially, place above a rear molar between the upper cheek and gum PRN; if pain is not relieved in 30 min, may repeat once. Max of 2 doses/pain episode; do not repeat additional doses for at least 4 h. Reevaluate maintenance opioid if there are > 4 episodes of breakthrough pain/d. Increase dose in a stepwise manner to next highest tablet strength (up to 800 mcg/dose) if several consecutive episodes require > 1 dose

 

Lozenge:

200 mcg initially, place between the cheek and lower gum PRN; if pain is not relieved in 30 min (15 min after completion of lozenge), may repeat dose once. Max of 2 doses/pain episode; do not repeat additional doses for at least 4 h. Increase dose to next highest lozenge strength if several consecutive episodes require > 1 lozenge. Do not use for > 4 breakthrough episodes/d

 

Intranasal:

100 mcg (1 spray) initially PRN; if pain is not relieved in 30 min, use alternative rescue medication (do not repeat intranasal dose for at least 2 h) and increase to a higher dose during the next episode; titrate dose in a stepwise manner (200 mcg, 300 mcg, 400 mcg, 600 mcg, 800 mcg) until effective dose is found (max, 800 mcg/dose). Do not use for > 4 breakthrough episodes/d

 

Sublingual tablet:

100 mcg sublingually initially PRN; if pain is not relieved in 30 min, may repeat dose once. Max of 2 doses/pain episode; do not repeat additional doses for at least 2 h. May titrate dose in a stepwise manner over consecutive episodes (200 mcg, 300 mcg, 400 mcg, 600 mcg, 800 mcg) until effective dose is found (max, 800 mcg/dose). Do not use for > 4 breakthrough episodes/d

 

Sublingual spray:

100 mcg (1 spray) sublingually initially PRN; if pain is not relieved in 30 min, may repeat dose once. Max of 2 doses/pain episode; do not repeat additional doses for at least 4 h. Titrate dose in a stepwise manner (200 mcg, 400 mcg, 600 mcg, 800 mcg, 1200 mcg, 1600 mcg) if several consecutive episodes require > 1 dose. Do not use for > 4 breakthrough episodes/d

 

IV or IM injection:

50-100 mcg IM or IV every 1-2 h PRN for recovery room pain; when used off-label for severe pain in critically ill patients, fentanyl can be administered intermittently (typical initial dosing, 25-35 mcg or 0.35-0.5 mcg/kg IV or IM every 30-60 min PRN) or via continuous infusion (typical initial dosing, 0.5-2 mcg/kg/h); when used during surgery as an adjunct to general anesthesia, dosing typically ranges from 1 to 20 mcg/kg IM or IV, depending on the procedure; 50-100 mcg IM may be administered 30-60 min before the procedure as premedication

Fentanyl products are not interchangeable; always start with the lowest dose, unless making one of the switches described below

 

When switching from the lozenge to the buccal tablet, start with a 100 mcg tablet if the patient used a 200 or 400 mcg lozenge; a 200 mcg tablet if the patient used a 600 or 800 mcg lozenge; and two 200 mcg tablets if the patient used a 1200 or 1600 mcg lozenge. For patients converting from lozenge strengths ≥ 600 mcg, titration can proceed in multiples of 200 mcg

 

When switching from the lozenge to the sublingual tablet, start with a 100 mcg tablet if the patient used a 200 mcg lozenge (may titrate in multiples of 100 mcg); a 200 mcg tablet if the patient used a lozenge strength of 400-1200 mcg (may titrate in multiples of 200 mcg); and a 400 mcg tablet if the patient used a 1600 mcg lozenge (may titrate in multiples of 400 mcg)

 

When switching from the lozenge to the sublingual spray, start with a 100 mcg spray if the patient used a 200 or 400 mcg lozenge (may titrate in multiples of 100 mcg); a 200 mcg spray if the patient used a lozenge strength of 600 or 800 mcg (may titrate in multiples of 200 mcg); and a 400 mcg spray if the patient used a 1200 or 1600 mcg lozenge (may titrate in multiples of 400 mcg)

 

Use caution in renal and hepatic impairment (monitor closely for adverse effects)

Hydromorphone

 

Tablet: 2 mg, 4 mg, 8 mg

 

Oral solution: 5 mg/5 mL

 

Rectal suppository: 3 mg

 

Solution for injection: 1 mg/mL, 2 mg/mL, 4 mg/mL, 10 mg/mL

 

CII

Pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

Tablets:

2-4 mg PO every 4-6 h initially, titrate to pain relief

 

Oral solution:

2.5-10 mg PO every 3-6 h PRN initially, titrate to pain relief

 

IV injection:

0.2-1 mg IV every 2-3 h initially, titrate to pain relief

 

IM/SC injection:

1-2 mg IM/SC every 2-3 h PRN initially, titrate to pain relief

 

Rectal suppository:

3 mg PR every 6-8 h, titrate to pain relief

High potency injection (10 mg/mL) should only be used in opioid-tolerant patients

 

IM administration is not recommended because it is painful and may have variable absorption; SC is preferred over IM

 

In renal and hepatic impairment, consider starting with 25-50% of the usual starting dose, depending on the degree of impairment

 

Take with food to decrease stomach irritation

 

 

Meperidine

 

Tablet: 50 mg, 100 mg

 

Oral solution: 50 mg/5 mL

 

Solution for injection: 25 mg/mL, 50 mg/mL, 75 mg/mL, 100 mg/mL

 

CII

Tablet and oral solution: Management of acute pain severe enough to require an opioid analgesic

 

Solution for injection: Management of acute pain severe enough to require an opioid analgesic; obstetrical analgesia; preoperative medication

Tablet and oral solution:

50-150 mg PO every 3-4 h PRN

 

IM/SC injection:

For pain, 50-150 mg IM/SC every 3-4 h PRN; for obstetric analgesia, 50-100 mg IM/SC, may repeat at 1-3 h intervals; for preoperative medication, 50-100 mg IM/SC 30-90 min before anesthesia

 

If given IV, use a reduced dose

 

Max for pain (any route): 150 mg/dose

 

Max for preoperative medication: 100 mg/dose

Prolonged use can lead to accumulation of normeperidine and increase risk of seizures

 

Avoid use in elderly patients and patients with renal dysfunction

 

Dilute oral solution in half a glass of water to prevent topical anesthesia of mucous membranes

Morphine

 

Tablet: 15 mg, 30 mg

 

Oral solution: 10 mg/5 mL, 20 mg/5 mL, 100 mg/5 mL

 

Rectal suppository: 5 mg, 10 mg, 20 mg, 30 mg

 

Solution for injection: 0.5 mg/mL, 1 mg/mL, 2 mg/mL, 4 mg/mL, 5 mg/mL, 8 mg/mL, 10 mg/mL, 25 mg/mL, 50 mg/mL

 

CII

Pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

Tablets and oral solution:

10-30 mg PO every 4 h PRN initially, titrate to pain relief

 

IV injection:

2.5-5 mg IV every 3-4 h PRN initially, titrate to pain relief (usual range, 5-15 mg every 4 h PRN)

 

IM/SC injection:

5-10 mg IM/SC every 4 h PRN initially, may increase up to 20 mg every 4 h PRN

 

Epidural injection:

2-3.75 mg as a single dose (usual range for opioid-naïve patients)

 

Epidural infusion:

0.2-0.4 mg/h for opioid-naïve patients; if using a continuous microinfusion device, the usual initial dose is 3.5-7.5 mg/24 h

 

Intrathecal injection:

0.1-0.3 mg as a single dose

 

Intrathecal infusion:

0.2-1 mg/24 h initially for opioid-naïve patients, usual maximum of 20 mg/24 h

 

Rectal suppository:

10-20 mg PR every 4 h

Concentrated oral morphine solution (100 mg/5 mL) should only be used in opioid-tolerant patients

 

Preservative-free injection solutions should be used for epidural and intrathecal administration

 

Active metabolites may accumulate in renal failure; avoid use if possible, particularly in patients with severe renal impairment (CrCl < 30 mL/min). Start with lower doses and titrate slowly in patients with renal or hepatic impairment

 

Take with food to decrease stomach irritation

Nalbuphine

 

Solution for injection: 10 mg/mL, 20 mg/mL

 

Not a controlled substance

Pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate; supplement to balanced anesthesia; preoperative and postoperative analgesia; analgesia during labor and delivery

For pain and labor-related pain:

10 mg IM, IV, or SC every 3-6 h PRN (max, 20 mg/dose or 160 mg/d in nonopioid-tolerant patients)

 

For use as an adjunct to anesthesia:

0.3-3 mg/kg IV over 10-15 min, then subsequent doses of 0.25-0.5 mg/kg IV PRN

 

Oxycodone

 

Tablet: 5 mg, 7.5 mg, 10 mg, 15 mg, 20 mg, 30 mg

 

Capsule: 5 mg

 

Oral solution: 5 mg/5 mL, 100 mg/5 mL

 

CII

Pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

5-15 mg PO every 4-6 h PRN initially, titrate to pain relief

In patients with hepatic impairment, start at 33% to 50% of the usual dose and titrate carefully

 

Use caution in CrCl < 60 mL/min, as serum concentrations may be increased; start with a low dose

 

Take with food to decrease stomach irritation

Oxymorphone

 

Tablet: 5 mg, 10 mg

 

CII

Pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

5-20 mg PO every 4-6 h PRN initially, titrate to pain relief

Contraindicated in moderate-to-severe hepatic impairment

 

Start at the lowest dose (ie, 5 mg) in patients with CrCl < 50 mL/min and patients with mild hepatic impairment

 

Administer on an empty stomach (1 h prior to or 2 h after eating)

Sufentanil

 

Sublingual tablet: 30 mcg

 

Solution for injection: 50 mcg/mL

 

CII

Sublingual tablet: Management of acute pain severe enough to require an opioid analgesic for which alternative treatments are inadequate

 

Solution for injection: Adjunct to anesthesia or primary anesthetic agent (IV); analgesia during labor and vaginal delivery (epidural, in combination with bupivacaine)

Sublingual tablet:

30 mcg sublingually every h PRN for up to 72 h; do not exceed 12 tablets/24 h

 

IV injection or infusion:

When used as an adjunct to general anesthesia, may use a total dose of 1-8 mcg/kg, depending on surgery length (not to exceed 1 mcg/kg/h of surgery); when used as the primary anesthetic agent, total doses per procedure may range from 8 to 30 mcg/kg

 

Epidural injection:

10-15 mcg mixed with 10 mL bupivacaine 0.125%; may repeat twice (up to a total of 3 doses) at intervals no less than 1 h

Sublingual sufentanil should only be used in a supervised health care setting

 

Use caution in renal and hepatic impairment (monitor closely for adverse effects)

Tapentadol

 

Tablet: 50 mg, 75 mg, 100 mg

 

CII

Management of acute pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

50-100 mg every 4-6 h; on the first day of therapy, a second dose may be given as soon as 1 h after the first dose if pain relief is not achieved

 

Max: 700 mg/d on the first day of therapy, then 600 mg/d

Avoid use in CrCl < 30 mL/min and severe hepatic impairment

 

In moderate hepatic impairment, reduce starting dose to 50 mg every 8 h (max, 150 mg/24 h)

Tramadol

 

Tablet: 50 mg, 100 mg

 

CIV

Pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

50-100 mg PO every 4-6 h PRN

 

If rapid onset of analgesia is not required, start with 25 mg PO daily and titrate in 25 mg increments every 3 d to 25 mg PO 4 times daily; then, increase total daily dose by 50 mg every 3 d up to 50 mg PO 4 times daily; after titration, 50-100 mg every 4-6 h can be administered PRN

 

Max: 400 mg/d

Do not exceed 300 mg/d in elderly patients (> 75 years old)

 

If CrCl < 30 mL/min, increase dosing interval to every 12 h and do not exceed 200 mg/d

 

In patients with severe hepatic impairment, use a dose of 50 mg PO every 12 h

Combination Products
APAP Combinations

Benzhydrocodone/APAP

 

Tablet: 4.08 mg/325 mg, 6.12 mg/325 mg, 8.16 mg/325 mg

 

CII

Short-term treatment of acute severe pain (no more than 14 d)

 

1-2 tablets PO every 4-6 h PRN

 

Max: 12 tablets/d

Use a low initial dose in renal/hepatic impairment

Codeine/APAP

 

Tablet: 15 mg/300 mg, 30 mg/300 mg, 60 mg/300 mg

 

 

Oral solution: 12 mg/120 mg per 5mL

 

CIII (tablets), CV (solution)

Mild-to-moderate pain

Tablets:

1-2 tablets PO every 4 h PRN

 

Oral solution:

15 mL PO every 4 h PRN

 

Max: 60 mg codeine per dose; 360 mg codeine and 4 g APAP per day

Take with full glass of water; may take with food to decrease stomach irritation

Hydrocodone/APAP

 

Tablet: 2.5 mg/325 mg, 5 mg/300 mg, 5 mg/325 mg, 7.5 mg/300 mg, 7.5 mg/325 mg, 10 mg/300 mg, 10 mg/325 mg

 

Oral solution: 7.5 mg/325 mg per 15 mL, 10 mg/325 mg per 15 mL

 

Oral elixir:

10 mg/300 mg per 15 mL

 

CII

Pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

Tablets containing 2.5-5 mg hydrocodone:

1-2 tablets PO every 4-6 h PRN

 

Tablets containing 7.5-10 mg hydrocodone:

1 tablet PO every 4-6 h PRN

 

Oral solution:

7.5-10 mg hydrocodone (15 mL) PO every 4-6 h PRN

 

Oral elixir:

11.25 mL PO every 4-6 h PRN

 

Max: depends on product, do not exceed 4 g APAP/d

Take with full glass of water; may take with food to decrease stomach irritation

Oxycodone/APAP

 

Tablet: 2.5 mg/300 mg, 2.5 mg/325 mg, 5 mg/300 mg, 5 mg/325 mg, 7.5 mg/300 mg, 7.5 mg/325 mg, 10 mg/300 mg, 10 mg/325 mg

 

CII

Pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

2.5-10 mg of oxycodone PO every 6 h PRN

 

Max: do not exceed 4 g APAP/d

Take with full glass of water; may take with food to decrease stomach irritation

Tramadol/APAP

 

Tablet: 37.5 mg/325 mg

 

CIV

Short-term (5 d or less) management of acute pain severe enough to require an opioid analgesic

2 tablets PO every 4-6 h PRN

 

Max: 8 tablets/24 h

Use not recommended in hepatic impairment

 

For CrCl < 30 mL/min, max dose is 2 tablets PO every 12 h

Ibuprofen Combinations

Hydrocodone/ibuprofen

 

Tablet: 5 mg/200 mg, 7.5 mg/200 mg, 10 mg/200 mg

 

CII

Short-term (< 10 d) management of acute pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

1 tablet PO every 4-6 h PRN

 

Max: 5 tablets/d

Use a low initial dose in renal impairment and severe hepatic impairment

 

Take with food to decrease stomach irritation

Oxycodone/ibuprofen

 

Tablet: 5 mg/400 mg

 

CII

Short-term (≤ 7 d) treatment of acute-to- moderate pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

1 tablet PO every 6 h PRN

 

Max: 4 tablets/24 h

Avoid use in advanced renal disease

 

Take with food to decrease stomach irritation

ASA Combinations

 

Oxycodone/ASA

 

Tablet: 4.8355 mg/325 mg

 

CII

Pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

1 tablet PO every 6 h PRN

 

Max: 12 tablets/24 h

Avoid use in severe renal impairment (CrCl < 10 mL/min) and hepatic impairment

 

Take with food to decrease stomach irritation

Multiple Drug Combinations

Dihydrocodeine/APAP/
caffeine

 

Capsule: 16 mg/320.5 mg/30 mg

 

Tablet: 16 mg/325 mg/30 mg

 

CIII

Pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

2 tablets or capsules PO every 4 h PRN

 

Max: 10 tablets or capsules/24 h

Use caution in renal/hepatic impairment

 

Take with full glass of water

Codeine/ASA/carisoprodol

 

Tablet: 16 mg/325 mg/
200 mg

 

CIII

Relief of discomfort associated with acute, painful musculoskeletal conditions

 

Use for short-term treatment (2-3 wk)

1-2 tablets PO 4 times daily

 

Max: 8 tablets/d

Avoid use in severe renal impairment (CrCl < 10 mL/min)

 

May take with food to decrease stomach irritation

Codeine/APAP/butalbital/caffeine

 

Capsule: 30 mg/
300 mg/50 mg/40 mg

 
30 mg/325 mg/50 mg/
40 mg

 

CIII

Relief of tension or muscle contraction headache

1-2 capsules PO every 4 h PRN

 

Max: 6 capsules/d

 

Codeine/ASA/butalbital/
caffeine

 

Capsule: 30 mg/
325 mg/50 mg/40 mg

 

CIII

Relief of tension or muscle contraction headache

1-2 capsules PO every 4 h

 

Max: 6 capsules/d

Avoid use in severe renal impairment (CrCl < 10 mL/min)

 

Take with a full glass of water

Other Combinations

 

Pentazocine/naloxone

 

Tablet: 50 mg/0.5 mg

 

CIV

Pain severe enough to require an opioid analgesic and for which alternative treatments are inadequate

1 tablet PO every 3-4 h

 

May increase up to 2 tablets PO every 3-4 h if needed

 

Max: 12 tablets/d

Use caution in hepatic impairment

 

CrCl 10-50 mL/min: reduce dose by 25%

 

CrCl < 10 mL/min: reduce dose by 50%

a Doses presented here are meant to provide a general guide for initial therapy. Patients who are opioid-tolerant may require higher doses. Use the lowest effective dose, and titrate up or down based on patient response.

Abbreviations: APAP, acetaminophen; ASA, aspirin; CrCl, creatinine clearance; FDA, US Food and Drug Administration; IM, intramuscular; IV, intravenous; PO, oral; PR, per rectum; PRN, as needed; SC, subcutaneous.

Long-Acting Opioid Analgesics and Extended-Release Formulations

The category of “long-acting opioids” encompasses two types of opioid analgesics: short-acting opioids that have been formulated into extended-release dosage forms, and opioids that are intrinsically long-acting. Long-acting opioids can be useful in the long-term treatment of chronic pain, because they allow for less frequent dosing and more consistent analgesia.9 Patients are recommended to initiate opioid treatment with a short-acting agent; they may then convert to a long-acting agent once an effective dose has been established, if desired.9-11 Some long-acting opioids are only indicated for use in opioid-tolerant patients.8,13

Morphine, fentanyl, hydrocodone, hydromorphone, tapentadol, tramadol, oxymorphone, and oxycodone are all short-acting opioids that are available in extended-release formulations.8 Intrinsically long-acting opioids include methadone, buprenorphine, and levorphanol. Tables 4 and 5 summarize indications and dosing for the long-acting opioids. The pharmacokinetic characteristics of long-acting drugs and formulations are summarized in Table 6.

Methadone is commonly used in the treatment of opioid dependence, but it can also be used to treat pain.2 Its oral bioavailability is variable, ranging from 36% to 100%.8 Its half-life is also highly variable, ranging from 8 to 59 hours or longer.8,13 It accumulates in tissues with repeated administration, so peak analgesic effects of a given dosing regimen may not be seen for 3 to 5 days.2,8,13 Its peak respiratory depressant effect occurs after its peak pharmacologic effect, so titration should be performed slowly and cautiously to avoid unintentional overdose.2

Buprenorphine has a long duration of action because it is slow to dissociate from MOR.1 Some formulations of buprenorphine are only indicated for treatment of opioid dependence.8 For pain treatment, buprenorphine is available as a transdermal patch, a buccal film, and a solution for injection. Bioavailability of the buccal film is 46% to 65%, and transdermal bioavailability is 15%.

Levorphanol acts similarly to morphine, but it is more potent than morphine and has a prolonged half-life.2 This causes it to accumulate with repeated dosing. Oral bioavailability is not known, but it is generally well-absorbed.8

Table 4. Long-Acting Opioid Analgesic Formulations.8,13
Generic Name and Formulations FDA Indication(s) Typical Adult Dosing for Pain Notes
Intrinsically Long-Acting Opioid Agents

Buprenorphinea

 

Buccal film (Belbuca): 75 mcg, 150 mcg, 300 mcg, 450 mcg, 600 mcg, 750 mcg, 900 mcg

 

Transdermal patch (Butrans): 5 mcg/h, 7.5 mcg/h, 10 mcg/h, 15 mcg/h, 20 mcg/h

 

Solution for injection (generic): 0.3 mg/mL

 

CIII

Management of pain severe enough to require daily, around-the-clock, long-term opioid treatment and for which alternative treatment options are inadequate (patch and buccal film); management of pain severe enough to require an opioid analgesic and for which alternate treatments are inadequate (solution for injection)

Buccal film:

Opioid-naïve patients:

75 mcg placed against the inside of the cheek daily or, if tolerated, every 12 h for at least 4 d, then increase to 150 mcg every 12 h

 

For all patients, titrate in increments of 150 mcg every 12 h no more frequently than every 4 d

 

Max: 900 mcg every 12 h

 

Transdermal patch:

Opioid-naïve patients:

5 mcg/h patch applied once every 7 d

 

For all patients, titrate in 5 mcg/h, 7.5 mcg/h, or 10 mcg/h increments (using no more than 2 patches) with a minimum titration interval of 72 h

 

Max: 20 mcg/h every 7 d

 

Solution for injection:

0.3 mg IV or IM; may repeat once if needed 30-60 min after the initial dose, then every 6-8 h PRN

 

Max: 0.3 mg/dose IV, 0.6 mg/dose IM

Buccal film:

In severe hepatic impairment, reduce starting dose and titration dose by 50%

 

Transdermal patch:

Avoid use in severe hepatic impairment

 

Apply to the upper outer arm, upper chest, upper back, or side of chest; rotate application sites (wait at least 21 d before using the same site)

 

Do not expose to external heat sources

Levorphanol

 

Tablet (generic): 2 mg, 3 mg

 

CII

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

Opioid-naïve patients:

1-2 mg PO every 6-8 h PRN; may increase to 3 mg PO every 6-8 h if needed

 

For all patients, titrate no more frequently than every 3 d

 

Reduce initial dose by 50% or more in debilitated elderly patients and patients with conditions or medications affecting respiratory reserve

 

Take with food to decrease stomach irritation

Methadonea

 

PO solution (generic): 5 mg/5 mL, 10 mg/5 mL, 10 mg/mL

 

Tablet (generic): 5 mg, 10 mg

 

Solution for injection (generic): 10 mg/mL

 

CII

Management of pain severe enough to require daily, around-the-clock, long-term opioid treatment and for which alternative treatment options are inadequate; detoxification and maintenance treatment of opioid use disorder (PO formulations); temporary treatment of opioid dependence in patients unable to take PO medication (solution for injection)

Tablets and PO solution:

Opioid-naïve patients:

2.5 mg PO every 8-12 h initially

 

For all patients, adjust dose no more frequently than every 3-5 d (some sources recommend titrating by no more than 10 mg/d every 5-7 d); up to 12 d may be necessary between dose increases for some patients; titrate to pain relief

 

Solution for injection:

Opioid-naïve or nonopioid-tolerant patients:

2.5-10 mg IV, IM, or SC every 8-12 h; titrate slowly to effect

Use caution in renal and hepatic impairment; for CrCl < 10 mL/min, consider reducing initial dose by 50-75%

 

Monitor closely for respiratory depression, especially within 24-72 h of therapy initiation or dose escalation

Opioids Available in Extended-Release Formulations

Fentanyl

 

Transdermal patch (Duragesic): 12 mcg/h, 25 mcg/h, 37.5 mcg/h, 50 mcg/h, 62.5 mcg/h, 75 mcg/h, 87.5 mcg/h, 100 mcg/h

 

CII

Management of pain in opioid-tolerant patientsb severe enough to require daily, around-the-clock opioid treatment and for which alternative treatment options are inadequate

Apply to the chest, back, flank, or upper arm. Change patch every 72 h (some patients may require a patch change every 48 h)

 

Do not titrate more frequently than every 3 d after the initial dose or every 6 d thereafter; short-acting agents can be used until analgesic efficacy is established

Rotate application sites

 

Avoid exposing the application site to external heat sources

 

Reduce initial dose by 50% in patients with mild-to-moderate renal or hepatic impairment; avoid use in severe renal or hepatic impairment

Hydrocodone

 

ER capsule (Zohydro ER):
10 mmg, 15 mg, 20 mg, 30 mg, 40 mg, 50 mg

 

ER tablet (Hysingla ER):c 20 mg, 30 mg, 40 mg, 60 mg, 80 mg, 100 mg, 120 mg

 

CII

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

ER capsule:

Nonopioid-tolerant patients:

10 mg PO every 12 h initially

 

For all patients, titrate by 10 mg every 12 h if needed every 3-7 d

 

ER tablet:

Nonopioid-tolerant patients:

20 mg PO every 24 h

 

For all patients, titrate by 10-20 mg as appropriate every 3-5 d

ER capsule:

In severe hepatic impairment, start with 10 mg every 12 h

 

Use a low initial dose in renal impairment

 

ER tablet:

In severe hepatic impairment or moderate-to-severe renal impairment, start with 50% of the initial dose

Hydromorphone

 

ER tablet (generic): 8 mg, 12 mg, 16 mg, 32 mg

Management of pain in opioid-tolerant patientsb severe enough to require daily, around-the-clock, long-term opioid treatment and for which alternative treatment options are inadequate

Converting from PO hydromorphone:

Give total daily hydromorphone dose PO once daily

 

Converting from other PO opioids:

Calculate total daily dose of the previous opioid(s) and use the conversion table in the prescribing information to calculate the approximate equivalent dose of hydromorphone; decrease calculated dose by 50% (round down if necessary) and give this dose PO once daily

 

Converting from fentanyl transdermal patch:

Calculate the total daily dose of hydromorphone using a conversion factor of 12 mg hydromorphone for each 25 mcg/h fentanyl transdermal patch; give 50% of the calculated dose (round down if necessary) PO daily beginning 18 h after removal of the fentanyl patch

 

Titration for all patients may occur in increments of 4-8 mg every 3-4 d

CrCl 40-60 mL/min: reduce starting dose to 50% of the usual starting dose

 

CrCl <40 mL/min: reduce starting dose to 25% of the usual starting dose; consider an alternative analgesic to permit more dosing flexibility

 

Moderate hepatic impairment: reduce starting dose to 25% of the usual starting dose

 

Avoid use in severe hepatic impairment

Morphine

 

ER capsule (Kadian): 10 mg, 20 mg, 30 mg, 40 mg, 50 mg, 60 mg, 80 mg, 100 mg, 200 mg

 

ER capsule, biphasic release (generic): 30 mg, 45 mg, 60 mg, 75 mg, 90 mg, 120 mg

 

ER tablet (MS Contin): 15 mg, 30 mg, 60 mg, 100 mg, 200 mg

 

ER tablet (MorphaBond ER):c 15 mg, 30 mg, 60 mg, 100 mg

 

CII

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

ER capsule:

Do not use in opioid-naïve patients

 

Nonopioid-tolerant patients:

30 mg PO every 24 h

 

Doses may be adjusted every 1-2 d for all patients

 

ER capsule, biphasic release:

Opioid-naïve or nonopioid-tolerant patients:

30 mg PO every 24 h

 

Doses may be adjusted in increments ≤ 30 mg every 3-4 d for all patients

 

Max: 1600 mg/d (due to fumaric acid in the formulation)

 

ER tablet:

Opioid- naïve patients:

15 mg PO every 8-12 h

 

Nonopioid-tolerant patients:

15 mg PO every 12 h (MS Contin) or every 8 or 12 h (MorphaBond ER)

 

Doses may be adjusted every 1-2 d for all patients

Active metabolites may accumulate in renal failure; avoid use if possible, particularly in patients with severe renal impairment (CrCl < 30 mL/min). Start with lower doses and titrate slowly in patients with renal or hepatic impairment

 

ER capsules may be opened and sprinkled over applesauce; Kadian may be administered via gastrostomy tube (do not administer via nasogastric tube)

Oxycodone

 

ER tablet (OxyContin):c
10 mg, 15 mg, 20 mg, 30 mg, 40 mg, 60 mg, 80 mg

 

ER capsule (Xtampza ER):c
9 mg, 13.5 mg, 18 mg, 27 mg, 36 mg

 

CII

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

 

ER tablet:

Opioid- naïve or nonopioid-tolerant patients:

10 mg PO every 12 h

 

ER capsule:

Opioid- naïve or nonopioid-tolerant patients:

9 mg PO every 12 h

 

Max (capsules only): 288 mg/d

 

Doses for all formulations may be adjusted in 25-50% increments no more frequently than every 1-2 d

Start with 33% to 50% of the recommended starting dose in hepatic impairment

 

Use caution in CrCl < 60 mL/min

 

ER tablet:

Start with 33-50% of the recommended starting dose in elderly, debilitated patients who are not opioid-tolerant

 

ER capsule:

Administer with food (capsules may be opened and sprinkled onto soft food or administered via nasogastric or gastrostomy tube)

Oxymorphone

 

ER tablet (generic): 5 mg, 7.5 mg, 10 mg, 15 mg, 20 mg, 30 mg, 40 mg

 

CII

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

Opioid- naïve or nonopioid-tolerant patients:

5 mg PO every 12 h

 

For all patients, titrate in increments of 5-10 mg every 12 h every 3-7 d

Contraindicated in moderate-to-severe hepatic impairment

 

For patients with mild hepatic impairment, elderly patients (≥ 65 years), and patients with CrCl < 50 mL/min

who were previously on another opioid, reduce calculated starting dose by 50%

 

Administer on an empty stomach (1 h prior to or 2 h after eating)

Tapentadol

 

ER tablet (Nucynta ER): 50 mg, 100 mg, 150 mg, 200 mg, 250 mg

 

CII

Management of chronic pain, including neuropathic pain associated with diabetic peripheral neuropathy, severe enough to require daily, around-the-clock, long-term opioid treatment and for which alternative treatment options are inadequate

Opioid-naïve or nonopioid-tolerant patients:

50 mg PO every 12 h

 

For all patients, titrate by no more than 50 mg/dose twice daily (100 mg/d) every 3 d within the range of 100-250 mg PO twice daily

 

Max: 500 mg/d

Use not recommended in severe renal or hepatic impairment

 

In moderate hepatic impairment, start with 50 mg PO every 24 h; max dose of 100 mg every 24 h

Tramadol

 

ER capsule, biphasic release (ConZip): 100 mg, 200 mg, 300 mg

 

ER dual-matrix tablet (generic): 100 mg, 200 mg, 300 mg

 

ER coated tablet (generic): 100 mg, 200 mg, 300 mg

 

CIV

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

Patients not currently taking tramadol:

100 mg PO daily

 

Converting from immediate release tramadol formulations:

When converting from PO immediate-release tramadol, determine the total daily tramadol dose and round down to the next lowest 100 mg increment

 

For all patients, titrate by 100 mg every 5 d

 

Max: 300 mg/d

Avoid use in severe hepatic impairment and CrCl < 30 mL/min

 

Take the ER coated tablet consistently with or without food

a Other formulations are available, but they are not indicated for the treatment of pain.
b The FDA considers a patient to be “opioid tolerant” if they have been taking, for 1 week or longer, at least 60 mg morphine per day, 25 mcg transdermal fentanyl per hour, 30 mg oral oxycodone per day, 8 mg oral hydromorphone per day, 25 mg oral oxymorphone per day, 60 mg oral hydrocodone per day, or an equianalgesic dose of another opioid.
c FDA-labeled as an abuse-deterrent formulation.

Abbreviations: APAP, acetaminophen; ASA, aspirin; ER, extended-release; FDA, US Food and Drug Administration; IM, intramuscular; IV, intravenous; PO, oral; PRN, as needed; SC, subcutaneous.
Table 5. Recommended Initial Fentanyl Patch Dosing Based on Daily Oral Morphine Equivalents.13
Oral Morphine Equivalents in a 24-hour Period Fentanyl Transdermal Dose
60-134 mg/d 25 mcg/h
135-224 mg/d 50 mcg/h
225-314 mg/d 75 mcg/h
315-404 mg/d 100 mcg/h
405-494 mg/d 125 mcg/h
495-584 mg/d 150 mcg/h
585-674 mg/d 175 mcg/h
675-764 mg/d 200 mcg/h
765-854 mg/d 225 mcg/h
855-944 mg/d 250 mcg/h
945-1034 mg/d 275 mcg/h
1035-1124 mg/d 300 mcg/h
Do not use this table to convert from transdermal fentanyl to other opioid analgesics; doing so will result in an overestimation of the new opioid dose and potential overdose.
Table 6. Pharmacokinetics of Long-Acting Opioid Analgesic Formulations.8,13,16
Generic Name Onset of Action Time to Peak Concentration Duration of Action/Half-Life
Intrinsically Long-Acting Opioid Agents
Buprenorphine buccal film NA 2.5-3 h Half-life, 27.6 h
Buprenorphine transdermal patch Quantifiable concentrations first detected after 17 h NA; steady state achieved by day 3 of first application Half-life, 26 h
Buprenorphine injection IM: ≥ 15 min IM: 1 h to peak effect IM: duration of action, 6-10 h

IV: half-life, 1.2-7.2 h
Levorphanol oral tablet 10-60 min 1 h Duration of action, 6-15 h; half-life, up to 30 h
Methadone oral tablet and solution 30-60 min 1-7.5 h (single dose)Peak analgesic effect occurs after 3-5 d of repeated administration Duration of action, 4-8 h for a single dose (increases to 22-48 h with repeated dosing); half-life, 8-59 h
Methadone IV injection 10-20 min Peak effect at 1-2 h Half-life, 8-59 h
Opioids Available in Extended-Release Formulations
Fentanyl transdermal patch 6-24 h 20-72 h Analgesia may last up to 12 h after patch removal; fentanyl levels decrease by 50% after about 17 h
Hydrocodone ER tablet NA 6-30 h Half-life, 7-9 h
Hydrocodone ER capsule NA 5 h Half-life, 8 h
Hydromorphone 6 h 12-16 h Systemic concentrations are sustained for 18-24 h post-dose; half-life, about 11 h
Morphine ER capsule NA 8-10 h Half-life, 11-13 h
Morphine ER capsule, biphasic release NA 30 min (contains immediate-release and ER component) Half-life, 24 h; concentrations maintained for 24 h
Morphine ER tablet NA 3-4 h NA
Oxycodone ER tablet NA 4-5 h Half-life of 4.5 h; duration of action ≤ 12 h
Oxycodone ER capsule NA 4-5 h Half-life, 5.6 h; duration of action, ≤ 12 h
Oxymorphone ER tablet NA 1-2 h Half-life, 9-11 h
Tapentadol ER tablet NA 3-6 h Half-life, 5-6 h
Tramadol ER capsule, biphasic release NA 5.9 h Half-life, 10 h
Tramadol ER dual-matrix tablet NA 4 h Half-life, 6.5 h
Tramadol ER coated tablet NA 12 h Half-life, 7.9 h
Abbreviations: ER, extended-release; IM, intramuscular; IV, intravenous; NA, not available.

Abuse-Deterrent Opioid Formulations

Most opioid analgesics have a high potential for misuse and abuse due to their euphoric effects and ability to cause physical dependence.17 Opioid misuse and abuse can lead to overdose, which can be fatal. In 2018, more than 67,300 people died of drug overdose in the United States: almost 70% of these overdose deaths involved an opioid drug.18 The number of overdose deaths involving opioids was 4 times higher in 2018 than in 1999. In light of this ongoing “opioid epidemic,” the FDA has encouraged the development of opioid formulations with reduced potential for abuse.19

Opioid abuse occurs in a variety of ways.20 These drugs may be swallowed whole, or they may be crushed and swallowed, snorted, smoked, or dissolved for injection. Abuse-deterrent opioid formulations are formulations with properties that have been shown to meaningfully deter abuse.19,20 These properties may make the drug more difficult to abuse, or make the drug less rewarding to abusers. Of note, abuse-deterrent formulations are not completely abuse-proof, and abuse-deterrent properties may not prevent addiction, overdose, or death. However, it is thought that the risk of abuse with abuse-deterrent formulations is lower than with nonabuse-deterrent formulations.

Abuse-deterrent properties may be conferred through the use of physical or chemical barriers, agonist/antagonist combinations, aversion, unique delivery systems, and/or new molecular entities and prodrugs.21 Physical barriers can help prevent the formulation from being chewed, crushed, cut, or ground, while chemical barriers can prevent the extraction of the opioid drug using common solvents. Both types of barriers can limit the amount of drug released when the formulation is manipulated, or change the physical form of the drug so that it is more difficult to abuse.20 Agonist/antagonist combinations include an antagonist to prevent abusers from achieving a “high.” The antagonist may be sequestered so that it is not released unless the product is inappropriately manipulated. Both of these strategies for abuse deterrence are limited by their inability to prevent the abuse of intact tablets.21

Aversion is another potential mechanism for abuse deterrence.20 This involves adding a substance to the drug’s formulation that produces an undesirable effect when the dosage form is manipulated or the medication is used at an inappropriately high dose. For example, the formulation may contain an ingredient that irritates the nasal mucosa, thus discouraging potential abusers from attempting to crush and snort the product. Aversion can help limit the abuse of intact tablets, and prevent abuse by chewing or crushing; however, it can also cause adverse effects in patients who are taking the tablets as intended and prevent legitimate dose increases.21 Additionally, if an abuser is highly motivated to abuse the drug, aversion techniques may not be sufficient to discourage them.

Certain delivery systems, such as depot injectable formulations or implants, can offer abuse resistance.20,21 It may be possible to extract the opioid from the delivery system, but it is much more difficult to manipulate these types of formulations. Another way to produce a formulation with abuse-deterrent properties would be to create a new molecular entity or prodrug with intrinsic properties that make it difficult to abuse.20 For example, the drug may require enzymatic activation, or it may penetrate the central nervous system more slowly, reducing the “high” that someone might feel if they abuse the drug.

In order to obtain abuse-deterrent labeling from the FDA, a drug manufacturer must perform studies that examine the potential for abuse.20 Premarket studies can fall into 1 of 3 categories: laboratory-based in vitro manipulation and extraction studies (Category 1), pharmacokinetic studies (Category 2), and clinical abuse potential studies (Category 3). The FDA generally recommends that manufacturers obtain data from all 3 types of studies to demonstrate that a formulation is likely to deter abuse in a meaningful way. However, in some cases, a particular category may not be relevant. Results of Category 1 studies can influence the design of Category 2 and 3 studies, and results of Category 2 studies can help determine the need for Category 3 studies.

The purpose of Category 1 studies is to see how easily the abuse-deterrent properties of the drug can be circumvented.20 These studies should consider ways in which abusers may try to manipulate the drug, as well as ways in which patients may manipulate the drug and inadvertently alter the rate or amount of drug released. Ease of mechanical manipulation (for example, by crushing, cutting, or grinding) should be examined, as well as the potential for dissolution and extraction of the opioid component. Other evaluations may depend on the anticipated route(s) of abuse: for example, if a product can be abused by smoking, the amount of drug produced by vaporization at various temperatures should be explored. For each type of study, the test product should be compared to other available products.

Category 2 studies are designed to compare the pharmacokinetics of the intact drug with the pharmacokinetics of the manipulated drug.20 The methods of manipulation used should reflect the methods that were examined in Category 1 studies and found to produce the greatest amount of drug release. These studies should also compare the pharmacokinetics of the intact and manipulated test product to the pharmacokinetics of intact and manipulated comparator formulations. Relevant pharmacokinetic parameters include maximum concentration (Cmax), time to maximum concentration (Tmax), area under the curve (AUC), and terminal elimination half-life (T1/2).

Category 3 studies assess the clinical abuse potential of a drug.20 These studies are also used to determine the FDA’s recommendation for drug scheduling. Typically, these studies are conducted in opioid-experienced recreational drug users who are familiar with the effects of the drug’s conventional formulation. The studies are usually designed as randomized, double-blind crossover studies, with a placebo control and a positive control. The test product is compared to the positive control, and the positive control is compared to placebo as a validation method. Different routes of abuse may be tested, depending on how the drug is likely to be abused in the real world. Likelihood of abuse is determined by how the subjects rate the drug’s effects. Visual analogue scales are used to measure “drug liking,” as well as good and bad effects of the drug and likelihood of taking the drug again.

Postmarketing studies (also referred to as Category 4 studies) are required for opioids with abuse-deterrent labeling.20 These studies assess the real-world impact of the formulation’s abuse-deterrent properties, evaluating its ability to produce meaningful reductions in abuse, misuse, and related adverse outcomes (such as addiction, overdose, and death). Formal Category 4 studies are observational in nature and may use a variety of data sources to assess overall and route-specific misuse and abuse. Rates of misuse and abuse with the abuse-deterrent formulation should be compared to rates of misuse and abuse seen with historically available and currently available nonabuse-deterrent formulations of the same drug. The FDA may require changes to the drug’s abuse-deterrent labeling, if postmarketing studies do not confirm that the formulation meaningfully deters abuse, or if studies show that the abuse-deterrent properties have caused abusers to shift to a riskier route of abuse.

The FDA has approved several products with specific “abuse-deterrent” labeling.8,19 Currently available products with abuse-deterrent labeling include OxyContin (extended-release oxycodone tablet), Xtampza ER (extended-release oxycodone capsule), Hysingla ER (extended-release hydrocodone tablet), and MorphaBond ER (extended-release morphine tablet).

OxyContin was the first opioid drug to receive FDA abuse-deterrent labeling.21 It is labeled as abuse-deterrent for intravenous and nasal routes of abuse.22 A previous formulation of OxyContin was implicated in a large number of opioid-related deaths and emergency department visits during the 2000s.23 This prompted a reformulation of the drug, and a new formulation with abuse-deterrent properties was approved in 2010. The reformulated version of OxyContin has physical properties that make it difficult to crush and snort or inject.21 It also resists attempts at dissolution and drug extraction: when it is put in water, it gradually forms a viscous gel, which is difficult to draw into a needle. A randomized, double-blind, placebo- and positive-controlled study in 27 recreational opioid users with a history of intranasal drug abuse found that overall drug liking and likelihood of taking the drug again were significantly reduced with crushed intranasal reformulated OxyContin vs crushed intranasal original OxyContin.24 Crushed reformulated OxyContin also had lower nasal tolerability. Pharmacokinetic studies performed in these patients found that, when crushed and taken intranasally, reformulated OxyContin had a decreased Cmax and an increased Tmax compared to the original formulation, with peak effects occurring at 1 to 2 hours after administration vs 30 minutes to 1 hour after administration.

Several observational postmarketing studies have been performed for reformulated OxyContin, but none of these postmarketing data have been included in the product labeling.21,25 Some postmarketing studies found reductions in OxyContin-specific abuse, diversion, overdose fatalities, and doctor shopping after the reformulation.21,26-28 One study noted that, while abuse rates of OxyContin decreased after reformulation, abuse rates of heroin and other opioids increased; this could indicate that some people who were previously abusing OxyContin simply switched to other more accessible opioid formulations.21,28 However, a recent study did not find an association between the reformulation of OxyContin and increased rates of new heroin use among patients who had abused OxyContin prior to the reformulation.29 A survey found that some people continued to abuse OxyContin via the oral route after the reformulation, and some people who had previously abused OxyContin via nonoral routes began abusing it via the oral route.30

Xtampza ER is a microsphere-in-capsule formulation of oxycodone labeled as abuse-deterrent for the intravenous, nasal, and oral routes.21,22 Relative to immediate-release oxycodone, it is more resistant to grinding, crushing, and extraction.21 It is also difficult to draw the melted capsule contents or the microspheres suspended in water into a needle. When crushed or chewed, the formulation maintains its extended-release pharmacokinetic profile.31-33 In a randomized, double-blind, placebo- and active-controlled study of 38 recreational opioid users, both chewed and intact Xtampza ER had lower drug liking scores and produced less of a subjective “high” compared to crushed immediate-release oxycodone taken orally.32 Similar results were found in a second study of 52 subjects.34 Another randomized, double-blind trial of 39 recreational opioid users found that Xtampza ER, given orally as an intact tablet or intranasally, produced lower drug liking scores and lower drug high scores than intranasal immediate-release oxycodone.35

Hysingla ER is a hydrocodone tablet with physical and chemical properties that make it difficult to crush, break, or dissolve.21 It retains some extended-release properties even if physically manipulated, and when put in water, it forms a viscous gel that is difficult to draw into a needle.36 It is labeled as abuse-deterrent for the intravenous, oral, and nasal routes.22 A randomized, double-blind trial of 40 recreational opioid users found that drug liking was lower for orally administered Hysingla ER compared to oral hydrocodone solution, whether Hysingla ER was taken intact, chewed, or ground into fine particles.37 A separate trial of 31 recreational opioid users compared hydrocodone powder to ground Hysingla ER and found that intranasal Hysingla ER produced less drug liking effects than intranasal hydrocodone powder.38 Hysingla ER also caused more nasal irritation.

MorphaBond ER (an extended-release morphine tablet) contains inactive ingredients that make it difficult to cut, crush, or break.21 The manipulated tablet also forms a gel when subjected to an aqueous environment. It is labeled as abuse-deterrent for the intravenous and nasal routes.22 MorphaBond ER retains some of its extended-release properties after manipulation.21 When crushed and taken intranasally, it has a lower Cmax than a nonabuse-deterrent extended-release morphine formulation (MS Contin). A randomized, double-blind, placebo- and active-controlled trial of 25 recreational opioid users found that MorphaBond ER had lower drug liking scores than crushed intranasal MS Contin when given orally (intact) or intranasally (crushed).39

Other abuse-deterrent opioid formulations have been approved; however, some of these have been discontinued, and others were never brought to market.8,22,25 Some opioid formulations underwent abuse deterrence testing but did not qualify for abuse-deterrent labeling.21 These include Oxaydo, an immediate-release formulation of oxycodone, and Zohydro ER, an extended-release hydrocodone formulation. In general, the approved abuse-deterrent opioids have used either physical/chemical barriers or agonist/antagonist combinations to deter abuse.

The impact of abuse-deterrent formulations is still largely unknown, and the evidence that does exist does not consistently demonstrate that these formulations reduce abuse.22 Patients may benefit more from an abuse-deterrent formulation if they are at high risk for abuse, or if they are at high risk of being targeted for diversion.40 A significant limitation of these formulations is that most are only available as branded products, which may mean higher costs and lack of insurance coverage.20-22,25

Conclusion

A variety of opioid drugs are available to treat pain. Opioid analgesics differ from one another in terms of potency, receptor activity, duration of action, indication, and dosage form. It is crucial for pharmacists to be aware of these differences, so that they can recommend an optimal drug and dosage form for a specific patient. Some opioid products have abuse-deterrent properties, but the clinical benefits of such formulations are currently unclear.

References

  1. Schumacher MA, Basbaum AI, Naidu RK. Opioid agonists & antagonists. In: Katzung BG, ed. Basic & Clinical Pharmacology. 14th ed. New York, NY: McGraw-Hill; 2018. http://accesspharmacy.mhmedical.com/content.aspx?bookid=2249&sectionid=175220393. Accessed May 8, 2020.
  2. Yaksh T, Wallace M. Opioids, analgesia, and pain management. In: Brunton LL, Hilal-Dandan R, Knollmann BC, eds. Goodman & Gilman's: The Pharmacological Basis of Therapeutics. 13th ed. New York, NY: McGraw-Hill; 2018. http://accesspharmacy.mhmedical.com/content.aspx?bookid=2189&sectionid=170269577. Accessed May 8, 2020.
  3. Fudin J, Perkins RJ, Lipman AG. Opioids. In: Cohen H, ed. Casebook in Clinical Pharmacokinetics and Drug Dosing. New York, NY: McGraw-Hill; 2015. http://accesspharmacy.mhmedical.com/content.aspx?bookid=1514&sectionid=88804586. Accessed May 8, 2020.
  4. Pathan H, Williams J. Basic opioid pharmacology: an update. Br J Pain. 2012;6(1):11-16.
  5. Emery MA, Eitan S. Members of the same pharmacological family are not alike: different opioids, different consequences, hope for the opioid crisis? Prog Neuropsychopharmacol Biol Psychiatry. 2019;92:428-449.
  6. Drewes AM, Jensen RD, Nielsen LM, et al. Differences between opioids: pharmacological, experimental, clinical and economical perspectives. Br J Clin Pharmacol. 2013;75(1):60-78.
  7. McPherson, ML. Demystifying Opioid Conversion Calculations: A Guide for Effective Dosing. 2nd ed. Bethesda, MD: American Society of Health-System Pharmacists; 2018.
  8. Clinical Pharmacology [database online]. Tampa, FL: Elsevier, Inc; 2020. https://www.clinicalkey.com/pharmacology/. Accessed March 14, 2020.
  9. Fine PG, Mahajan G, McPherson ML. Long-acting opioids and short-acting opioids: appropriate use in chronic pain management. Pain Med. 2009;10 Suppl 2:S79-S88.
  10. Dowell D, Haegerich TM, Chou R. CDC guideline for prescribing opioids in chronic pain—United States, 2016. JAMA. 2016;315(15):1624-1645.
  11. Manchikanti L, Kaye AM, Knezevic NN, et al. Responsible, safe, and effective prescription of opioids for chronic non-cancer pain: American Society of Interventional Pain Physicians (ASIPP) guidelines. Pain Physician. 2017;20(2S):S3-S92.
  12. Herndon CM, Ray JB, Kominek CM. Pain management. In: DiPiro JT, Yee GC, Posey L, Haines ST, Nolin TD, Ellingrod V, eds. Pharmacotherapy: A Pathophysiologic Approach. 11th ed. New York, NY: McGraw-Hill; 2019. http://accesspharmacy.mhmedical.com/content.aspx?bookid=2577&sectionid=226724502. Accessed May 8, 2020.
  13. Facts & Comparisons [database online]. St. Louis, MO: Wolters Kluwer; 2020. https://fco.factsandcomparisons.com/lco/action/home. Accessed March 14, 2020.
  14. Li JX. Combining opioids and non-opioids for pain management: current status. Neuropharmacology. 2019;158:107619. doi: 10.1016/j.neuropharm.2019.04.025.
  15. FDA drug safety communication: prescription acetaminophen products to be limited to 325 mg per dosage unit; boxed warning will highlight potential for severe liver failure. U.S. Food and Drug Administration website. https://www.fda.gov/drugs/drug-safety-and-availability/fda-drug-safety-communication-prescription-acetaminophen-products-be-limited-325-mg-dosage-unit. Updated February 7, 2018. Accessed May 8, 2020.
  16. Morphine sulfate capsule, extended release [package insert]. Parsippany, NJ: Actavis Pharma, Inc; 2019.
  17. Walsh SL, Babalonis S. The abuse potential of prescription opioids in humans—closing in on the first century of research. Curr Top Behav Neurosci. 2017;34:33-58.
  18. Understanding the epidemic. Centers for Disease Control and Prevention website. https://www.cdc.gov/drugoverdose/epidemic/index.html. Updated March 19, 2020. Accessed May 8, 2020.
  19. Abuse-deterrent opioid analgesics. US Food and Drug Administration website. https://www.fda.gov/drugs/postmarket-drug-safety-information-patients-and-providers/abuse-deterrent-opioid-analgesics. Updated June 11, 2019. Accessed May 8, 2020.
  20. Abuse-deterrent opioids—evaluation and labeling: guidance for industry. US Food and Drug Administration website. https://www.fda.gov/media/84819/download. Updated April 2015. Accessed May 8, 2020.
  21. Pergolizzi JV Jr, Raffa RB, Taylor R Jr, Vacalis S. Abuse-deterrent opioids: an update on current approaches and considerations. Curr Med Res Opin. 2018;34(4):711-723.
  22. Litman RS, Pagan OH, Cicero TJ. Abuse-deterrent opioid formulations. Anesthesiology. 2018;128(5):1015-1026.
  23. Timeline of selected FDA activities and significant events addressing opioid misuse and abuse. US Food and Drug Administration website. https://www.fda.gov/drugs/information-drug-class/timeline-selected-fda-activities-and-significant-events-addressing-opioid-misuse-and-abuse. Updated December 20, 2019. Accessed May 8, 2020.
  24. Harris SC, Perrino PJ, Smith I, et al. Abuse potential, pharmacokinetics, pharmacodynamics, and safety of intranasally administered crushed oxycodone HCl abuse-deterrent controlled-release tablets in recreational opioid users. J Clin Pharmacol. 2014;54(4):468-477.
  25. Carinci AJ. Abuse-deterrent opioid analgesics: a guide for clinicians. Pain Manag. 2020;10(1):55-62.
  26. Coplan PM, Chilcoat HD, Butler SF, et al. The effect of an abuse-deterrent opioid formulation (OxyContin) on opioid abuse-related outcomes in the postmarketing setting. Clin Pharmacol Ther. 2016;100(3):275-286.
  27. Severtson SG, Ellis MS, Kurtz SP, et al. Sustained reduction of diversion and abuse after introduction of an abuse deterrent formulation of extended release oxycodone. Drug Alcohol Depend. 2016;168:219-229.
  28. Coplan PM, Kale H, Sandstrom L, et al. Changes in oxycodone and heroin exposures in the National Poison Data System after introduction of extended-release oxycodone with abuse-deterrent characteristics. Pharmacoepidemiol Drug Saf. 2013;22(12):1274-1282.
  29. Wolff C, Dowd WN, Ali MM, et al. The impact of the abuse-deterrent reformulation of extended-release OxyContin on prescription pain reliever misuse and heroin initiation. Addict Behav. 2020;105:106268. doi: 10.1016/j.addbeh.2019.106268.
  30. Cicero TJ, Ellis MS. Abuse-deterrent formulations and the prescription opioid abuse epidemic in the United States: lessons learned from OxyContin. JAMA Psychiatry. 2015;72(5):424-430.
  31. Gudin J, Levy-Cooperman N, Kopecky EA, Fleming AB. Comparing the effect of tampering on the oral pharmacokinetic profiles of two extended-release oxycodone formulations with abuse-deterrent properties. Pain Med. 2015;16(11):2142-2151.
  32. Kopecky EA, Fleming AB, Levy-Cooperman N, et al. Oral human abuse potential of oxycodone DETERx® (Xtampza®ER). J Clin Pharmacol. 2017;57(4):500-512.
  33. Kopecky EA, Fleming AB, Noonan PK, et al. Impact of physical manipulation on in vitro and in vivo release profiles of oxycodone DETERx®: an extended-release, abuse-deterrent formulation. J Opioid Manag. 2014;10(4):233-246.
  34. Meske D, Kopecky EA, Passik S, Shram MJ. Evaluation of the oral human abuse potential of oxycodone DETERx® formulation (Xtampza® ER). J Opioid Manag. 2018;14(5):359-372.
  35. Webster LR, Kopecky EA, Smith MD, Fleming AB. A randomized, double-blind, double-dummy study to evaluate the intranasal human abuse potential and pharmacokinetics of a novel extended-release abuse-deterrent formulation of oxycodone. Pain Med. 2016;17(6):1112-1130.
  36. Hysingla ER [package insert]. Stamford, CT: Purdue Pharma LP; 2019.
  37. Harris SC, Cipriano A, Colucci SV, et al. Oral abuse potential, pharmacokinetics, and safety of once-daily, single-entity, extended-release hydrocodone (HYD) in recreational opioid users. Pain Med. 2017;18(7):1278-1291.
  38. Harris SC, Cipriano A, Colucci SV, et al. Intranasal abuse potential, pharmacokinetics, and safety of once-daily, single-entity, extended-release hydrocodone (HYD) in recreational opioid users. Pain Med. 2017;18(7):1278-1291.
  39. Webster LR, Pantaleon C, Shah MS, et al. A randomized, double-blind, double-dummy, placebo-controlled, intranasal drug liking study on a novel abuse-deterrent formulation of morphine-morphine ARER. Pain Med. 2017;18(7):1303-1313.
  40. Lee YH, Brown DL, Chen HY. Current impact and application of abuse-deterrent opioid formulations in clinical practice. Pain Physician. 2017;20(7):E1003-E1023. 

Back to Top