A Comprehensive Monograph on Sufentanil (DrugBank ID: DB00708): Pharmacology, Clinical Use, and Risk Management
Section 1: Executive Summary
Sufentanil is a potent, small-molecule synthetic opioid analgesic belonging to the 4-anilidopiperidine class of drugs. As a thienyl derivative of its parent compound, fentanyl, it represents one of the most powerful opioids utilized in modern clinical practice.[1] Its pharmacological profile is defined by an exceptionally high affinity and selectivity for the μ-opioid receptor, resulting in an analgesic potency estimated to be 5 to 10 times that of fentanyl and 500 to 1,000 times that of morphine.[1] This extreme potency, combined with high lipophilicity, facilitates a rapid onset of action, making it a cornerstone of anesthetic and analgesic practice in specific, high-acuity medical environments.[4]
The clinical applications of sufentanil are highly specialized and reflect its unique pharmacodynamic and pharmacokinetic properties. It is primarily administered intravenously as an analgesic adjunct to maintain balanced general anesthesia or as a primary anesthetic agent, particularly in major surgical procedures such as cardiovascular and neurosurgery, where its ability to ensure hemodynamic stability by attenuating the sympathetic stress response is highly valued.[3] Additionally, it is used via the epidural route, typically in combination with a local anesthetic like bupivacaine, to provide effective analgesia during labor and vaginal delivery.[5] A significant innovation in its delivery is the development of a sublingual tablet formulation (Dsuvia®, Dzuveo®), approved for the management of severe, acute pain exclusively within certified, medically supervised healthcare settings.[6]
The clinical utility of sufentanil is inextricably linked to its significant risk profile. Its profound potency means that the margin for dosing error is minimal, and its primary adverse effect, respiratory depression, can be rapid, severe, and potentially fatal.[3] Consequently, sufentanil is subject to stringent regulatory controls globally. In the United States, it is classified as a Schedule II controlled substance by the Drug Enforcement Administration (DEA), signifying a high potential for abuse, which may lead to severe psychological or physical dependence.[8] The unique risks associated with the sublingual tablet formulation, particularly the potential for accidental exposure and overdose, prompted the U.S. Food and Drug Administration (FDA) to mandate a Risk Evaluation and Mitigation Strategy (REMS) program. This program restricts its distribution and administration to a closed system of certified healthcare facilities, effectively prohibiting its use in outpatient or home settings.[10]
In conclusion, sufentanil occupies a critical but narrow niche in pharmacotherapy. Its high therapeutic index and unique ability to provide profound analgesia with hemodynamic stability make it invaluable for specific complex patient populations and procedures.[4] However, its safe and effective use is entirely dependent on a framework of expert clinical knowledge, strict administration protocols, and robust systemic risk-management strategies. The medication itself is a testament to the principle that the benefits of a powerful therapeutic agent can only be realized when the system of care is meticulously designed to mitigate its inherent risks.
Section 2: Identification and Physicochemical Profile
This section establishes the fundamental chemical, physical, and regulatory identity of sufentanil. The provision of standardized identifiers is crucial for ensuring accuracy and consistency in clinical practice, academic research, pharmacovigilance, and regulatory oversight on a global scale.
2.1 Nomenclature and Synonyms
To prevent ambiguity, sufentanil is identified by a standardized set of names and codes across various chemical and medical databases.
- Generic Name: Sufentanil [3]
- Systematic (IUPAC) Name: N-[4-(methoxymethyl)-1-(2-thiophen-2-ylethyl)piperidin-4-yl]-N-phenylpropanamide [8]
- Chemical Synonyms: The compound is also known by several synonyms, reflecting its chemical structure and development history. These include: N-(4-(Methoxymethyl)-1-(2-(2-thienyl)ethyl)-4-piperidinyl)-N-phenylpropanamide, N-(4-(Methoxymethyl)-1-(2-(2-thienyl)ethyl)-4-piperidyl)propionanilide, Sufentanilo, and Sufentanilum.[3] Its original research code from Janssen Pharmaceutica is R-30730.[6]
2.2 Chemical Structure and Properties
Sufentanil is a synthetic organic small molecule with a well-defined structure that dictates its pharmacological activity.[3]
- Chemical Formula: C22H30N2O2S [1]
- Molecular Weight: The average molecular weight is 386.55 g·mol⁻¹, with a monoisotopic mass of 386.202798904 Da.[1]
- Structural Class: Chemically, sufentanil is classified as a complex molecule belonging to several functional groups. It is an anilide, a member of piperidines, a member of thiophenes, and an ether.[8] More specifically, it is a thienyl derivative of fentanyl, which is a 4-anilidopiperidine.[2]
- Key Structural Features: Sufentanil was first synthesized in 1974 by modifying the structure of its parent compound, fentanyl.[1] The key structural differences are:
- The replacement of the phenyl ring attached to the propionamide group in fentanyl with a bioisosteric thiophene ring.[1]
- The addition of a methoxymethyl group at the 4-position of the piperidine ring. This modification is believed to significantly increase potency while potentially reducing the duration of action.[1]
- Physicochemical Properties: Sufentanil exists as a solid, specifically as crystals from petroleum ether, with a melting point reported to be approximately 97 °C (207 °F).[1] A defining physicochemical property is its high lipophilicity (fat-solubility), which is greater than that of fentanyl and is a primary determinant of its rapid transit across the blood-brain barrier and its overall pharmacokinetic profile.[4]
The extensive catalog of unique identifiers for sufentanil across numerous international databases is not merely an administrative formality. This complex web of codes is a direct reflection of the medication's status as a globally significant, high-risk therapeutic agent. The existence of distinct identifiers in chemical registries (CAS), comprehensive drug databases (DrugBank), therapeutic classification systems (ATC), and legal control frameworks (DEA) illustrates the multifaceted nature of its management. A simpler, lower-risk compound would not necessitate such a dense network of tracking codes. This infrastructure is essential for coordinated international efforts in pharmacovigilance to monitor adverse events, for law enforcement to control illicit manufacturing and diversion, and for the scientific community to conduct and compare research findings unambiguously. Therefore, this list of identifiers serves as a fingerprint of sufentanil's complex role in medicine and the global systems required to handle it safely.
Identifier | Value | Source(s) |
---|
DrugBank ID | DB00708 | 3 |
CAS Number | 56030-54-7 | 1 |
PubChem CID | 41693 | 1 |
UNII | AFE2YW0IIZ | 1 |
DEA Code Number | 9740 | 8 |
ATC Code | N01AH03 | 1 |
Chemical Formula | C22H30N2O2S | 1 |
Average Molecular Weight | 386.55 g·mol⁻¹ | 1 |
Table 2.1: Key Chemical and Physical Identifiers for Sufentanil | | |
Section 3: Comprehensive Pharmacological Profile
The clinical utility and risk profile of sufentanil are direct consequences of its interactions with biological systems. This section provides a detailed analysis of its mechanism of action at the molecular level, its systemic effects on the body (pharmacodynamics), and its absorption, distribution, metabolism, and excretion (pharmacokinetics).
3.1 Mechanism of Action: A Highly Selective μ-Opioid Receptor Agonist
Sufentanil exerts its effects primarily by acting as a potent and highly selective agonist at the μ-opioid receptor, a class of G-protein coupled receptors (GPCRs) widely distributed throughout the central and peripheral nervous systems.[3] Its affinity for the μ-receptor is approximately 100 times greater than its affinity for the δ-receptor.[4] While its primary action is at the μ-receptor, at higher concentrations, it can also bind to and activate δ- and κ-opioid receptors.[3]
The binding of sufentanil to the μ-opioid receptor initiates a cascade of intracellular signaling events that ultimately modulate neuronal activity:
- G-Protein Activation: Receptor binding facilitates the exchange of guanosine diphosphate (GDP) for guanosine triphosphate (GTP) on the associated inhibitory G-protein (Gi/Go) complex.[3]
- Inhibition of Adenylate Cyclase: The activated G-protein inhibits the enzyme adenylate cyclase, leading to a decrease in the intracellular concentration of the second messenger cyclic adenosine monophosphate (cAMP).[3]
- Modulation of Ion Channels: The signaling cascade also leads to the closure of presynaptic N-type voltage-gated calcium channels and the opening of postsynaptic G-protein-coupled inwardly-rectifying potassium (GIRK) channels.[3]
- Neuronal Inhibition: The combination of these effects—reduced calcium influx presynaptically and potassium efflux postsynaptically—results in membrane hyperpolarization and reduced neuronal excitability. This inhibits the release of key nociceptive (pain-signaling) neurotransmitters, including substance P, glutamate, and calcitonin gene-related peptide (CGRP), from the terminals of primary afferent neurons in the spinal cord and other CNS regions.[3]
3.2 Pharmacodynamics: Systemic Effects and Clinical Manifestations
The activation of opioid receptors in various organ systems produces a wide range of physiological effects, which constitute both the therapeutic actions and the adverse effects of sufentanil.
- Central Nervous System (CNS):
- Analgesia and Sedation: The principal therapeutic effects are profound analgesia, achieved by increasing the pain threshold and altering the perception of pain, and sedation.[3]
- Anesthesia: At intravenous doses of 8 mcg/kg or higher, sufentanil induces a deep level of anesthesia and hypnosis, sufficient for major surgery without the need for additional anesthetic agents. This state is characterized by specific alterations in electroencephalogram (EEG) patterns.[3]
- Respiratory Depression: Sufentanil produces a dose-dependent depression of the brainstem respiratory centers, reducing their responsiveness to carbon dioxide. This is the most significant and life-threatening adverse effect, and the respiratory depression may persist longer than the analgesic effect.[3]
- Other CNS Effects: It also depresses the cough reflex, causes miosis (pupillary constriction) via stimulation of the Edinger-Westphal nucleus, and can induce skeletal muscle rigidity, particularly of the truncal muscles, with rapid, high-dose intravenous administration. This rigidity is thought to result from effects on the substantia nigra and striate nucleus.[3]
- Cardiovascular and Hemodynamic Effects:
- Bradycardia: Sufentanil frequently causes a decrease in heart rate (bradycardia), likely due to a central vagotonic effect.[3]
- Vasodilation and Hypotension: It produces peripheral vasodilation, which can lead to orthostatic hypotension and syncope.[3]
- Hemodynamic Stability: A key clinical advantage of sufentanil, particularly in high-risk surgery, is its ability to produce a dose-related attenuation of the catecholamine (e.g., norepinephrine) stress response to surgical stimuli. At doses of 25-30 mcg/kg, it provides excellent hemodynamic stability and preserves a favorable myocardial oxygen supply-demand balance, making it a preferred agent for cardiovascular surgery.[5]
- Gastrointestinal (GI) and Other Systemic Effects:
- Reduced Motility: Sufentanil increases smooth muscle tone in the antrum of the stomach and the duodenum and decreases propulsive peristaltic waves in the colon. This slows digestion and transit, leading to constipation.[3]
- Biliary Effects: It can cause spasm of the Sphincter of Oddi, increasing pressure in the biliary tract. This necessitates caution in patients with biliary tract disease or pancreatitis.[3] It may also lead to temporary elevations in serum amylase.[3]
3.3 Pharmacokinetics: Absorption, Distribution, Metabolism, and Excretion (ADME)
The clinical behavior of sufentanil is governed by its movement into, through, and out of the body. Its pharmacokinetic profile is characterized by rapid onset, complex distribution, and significant variability among different patient populations.
- Absorption:
- Intravenous (IV): Administration by this route results in 100% bioavailability and an immediate onset of action.[1]
- Sublingual (SL): The bioavailability of a single sublingual tablet is approximately 53%. However, this value decreases to around 35% with repeated hourly dosing, likely due to local vasoconstriction or changes in absorption dynamics. After a single 30 mcg sublingual dose, plasma concentrations reach the analgesic threshold within 30 minutes and peak at approximately 1 hour.[1]
- Epidural: Following epidural administration, systemic absorption is minimal, with maternal and neonatal plasma concentrations often remaining at or below the lower limit of detection (0.05-0.1 ng/mL).[3] The primary effect is mediated by local action on spinal opioid receptors.[7]
- Distribution:
- Compartmental Model: The pharmacokinetics of intravenous sufentanil are best described by a three-compartment model, characterized by a rapid initial distribution phase (half-time ~1.4 minutes), a redistribution phase (half-time ~17.1 minutes), and a slower terminal elimination phase.[4]
- Lipophilicity and CNS Entry: Sufentanil's high lipophilicity allows it to rapidly cross the blood-brain barrier and equilibrate with its site of action in the CNS. The blood-effect-site equilibration half-life (t1/2ke0) is approximately 6.2 minutes, which accounts for its rapid onset of clinical effect.[4]
- Volume of Distribution (Vd): The volume of distribution at steady state (Vdss) in adults is approximately 1.7 L/kg, which is smaller than that of fentanyl.[4] The central volume of distribution is about 14 L.[3]
- Plasma Protein Binding: Sufentanil is extensively bound to plasma proteins, primarily alpha-1-acid glycoprotein (AAG). The bound fraction is approximately 91-93% in healthy adults. This binding is lower in neonates (~79%) due to their lower AAG concentrations.[2]
- Metabolism:
- Site and Pathway: The primary sites of biotransformation are the liver and the small intestine.[3] Metabolism occurs predominantly via the Cytochrome P450 3A4 (CYP3A4) enzyme system.[10]
- Metabolic Reactions: The main metabolic pathways are oxidative N-dealkylation and O-dealkylation, which convert sufentanil into a number of inactive metabolites.[3]
- Excretion:
- Route: The inactive metabolites are primarily excreted in the urine. Approximately 80% of an administered dose is excreted within 24 hours.[3]
- Unchanged Drug: Only a very small fraction, about 2%, of the dose is eliminated as unchanged sufentanil.[3]
- Elimination Half-life (t1/2): The terminal elimination half-life following IV administration in adults is approximately 164 minutes (2.7 hours).[1] This parameter is highly dependent on patient age, being significantly longer in neonates and shorter in young children compared to adults.[3]
The clinical utility of sufentanil is a direct consequence of a causal chain that begins with its chemical structure. Its high lipophilicity allows it to rapidly penetrate the blood-brain barrier, resulting in a very fast onset of action, making it ideal for anesthetic induction and for blunting intense, transient stimuli like tracheal intubation. This same property, however, also causes it to redistribute quickly from the central compartment (blood and brain) into peripheral tissues like fat and muscle. Following a single bolus injection, this rapid redistribution leads to a swift decline in plasma concentration and thus a short duration of clinical effect. In contrast, during a prolonged infusion, these peripheral compartments become saturated with the drug. When the infusion is stopped, the drug slowly leaches back into the plasma to be metabolized, a process governed by the much longer terminal elimination half-life. This phenomenon is described by the concept of context-sensitive half-time, which explains why recovery from a long sufentanil infusion is significantly slower than recovery from a single bolus. This complex kinetic profile dictates its clinical dosing strategies: single boluses are used for rapid, short-term effects, while carefully titrated continuous infusions are used for maintenance of anesthesia, with the understanding that the time to recovery is dependent on the duration of the infusion.
Furthermore, the high degree of plasma protein binding is a critical safety consideration, particularly in the neonatal population. In adults, sufentanil is approximately 93% bound to plasma proteins, mainly AAG, meaning only about 7% of the drug is in its free, pharmacologically active form. Neonates, however, have significantly lower circulating levels of AAG, resulting in a lower protein-bound fraction of about 79%. This seemingly small difference means that for the same total plasma concentration, a neonate has a substantially higher concentration of free, active sufentanil. This increased availability of active drug, combined with the neonate's immature hepatic enzyme systems and reduced clearance, dramatically increases their sensitivity to the drug's effects and elevates the risk of profound, prolonged respiratory depression and overdose. Therefore, the clinical guidance to use sufentanil with extreme caution and significantly reduced doses in neonates is not merely a general precaution but is a necessary adjustment based on these specific, quantifiable pharmacokinetic differences.
Parameter | Neonates | Infants/Children (2-8 yrs) | Adolescents/Adults | Source(s) |
---|
Elimination Half-life (IV) | ~434 ± 160 min | ~97 ± 42 min | ~164 ± 22 min | 3 |
Clearance | Approx. half of adult value | 30.5 ± 8.8 mL/min/kg | 12.7 ± 0.8 mL/min/kg | 3 |
Plasma Protein Binding | ~79% | Not specified, likely approaching adult levels | ~91-93% | 3 |
Table 3.1: Key Pharmacokinetic Parameters of Sufentanil across Patient Populations | | | | |
Section 4: Clinical Applications and Dosing Regimens
The translation of sufentanil's potent pharmacology into safe and effective clinical practice requires a nuanced understanding of its approved indications and highly specific dosing strategies. Administration is restricted to trained personnel in settings equipped to manage its powerful effects.
4.1 Approved Indications and Therapeutic Roles
Sufentanil is approved for use via intravenous, epidural, and sublingual routes, each with distinct clinical objectives.
- Intravenous Administration (Adults and Pediatrics):
- Analgesic Adjunct: It is indicated as an analgesic component to supplement and maintain balanced general anesthesia in patients who are intubated and receiving mechanical ventilation.[5]
- Primary Anesthetic Agent: At higher doses, it is approved as a primary anesthetic agent, administered with 100% oxygen, for the induction and maintenance of anesthesia. This indication is typically reserved for patients undergoing major surgical procedures, such as cardiovascular or neurosurgical operations, where profound anesthesia and hemodynamic stability are paramount, or when extended postoperative ventilation is anticipated.[3]
- Epidural Administration (Labor and Delivery):
- Obstetric Analgesia: It is indicated for epidural administration as an analgesic, almost always combined with a low concentration of a local anesthetic such as bupivacaine, to provide pain relief during labor and vaginal delivery.[5] The combination potentiates the analgesic effect, allowing for lower doses of the local anesthetic and thereby reducing motor blockade.[7]
- Sublingual Administration (Adults):
- Acute Pain Management: The sublingual tablet formulation (Dsuvia®) is indicated for the management of acute pain that is severe enough to require an opioid analgesic in adult patients within certified, medically supervised healthcare settings. These settings include hospitals, surgical centers, and emergency departments. This formulation has several key limitations: it is not for home use, its use has not been studied beyond 72 hours, and it is reserved for patients for whom alternative treatments are inadequate.[6]
4.2 Dosage and Administration
The administration of sufentanil demands meticulous attention to dosing, which must be individualized for each patient.
- General Principles of Administration:
- Individualization: Dosage must be tailored based on factors including age, body weight (with lean body weight recommended for obese patients), physical status (ASA classification), underlying pathological conditions, concomitant drug use, the type of anesthesia planned, and the nature of the surgical procedure.[4]
- Qualified Personnel: Sufentanil should only be administered by individuals specifically trained in the use of intravenous and epidural anesthetics and in the management of the respiratory effects of potent opioids, including airway management and resuscitation.[20]
- Availability of Reversal Agents: An opioid antagonist (e.g., naloxone), along with resuscitative equipment and oxygen, must be immediately available whenever sufentanil is used.[20]
- Accurate Measurement: For administering small intravenous or epidural volumes accurately, the use of a tuberculin syringe or an equivalent precision device is recommended.[20]
The wide spectrum of recommended doses for sufentanil is not arbitrary but directly mirrors its dose-dependent pharmacodynamic effects. The various dosing tiers outlined in clinical guidelines represent a practical map of the drug's dose-response curve. At lower concentrations, such as intravenous doses up to 8 mcg/kg, sufentanil primarily acts on nociceptive pathways to provide potent analgesia. As the dose and corresponding CNS concentration increase to levels of 8 mcg/kg or higher, the drug's effects expand to include profound sedation and hypnosis, sufficient to induce and maintain a state of general anesthesia. At the highest end of the therapeutic range, from 25 to 30 mcg/kg, sufentanil provides the additional, highly valued benefit of profound sympathetic blockade. This effect attenuates the release of stress hormones like catecholamines, leading to exceptional hemodynamic stability, which is why this dosage is specifically employed in high-stress procedures like cardiac surgery. Therefore, clinicians titrate the dose of sufentanil not simply to achieve "more" effect, but to target qualitatively different and specific clinical states—analgesia, hypnosis, or sympathetic blockade—as required by the surgical context and patient's condition.
Indication/Procedure | Patient Population | Route | Induction Dose | Maintenance Dose/Regimen | Key Clinical Notes |
---|
Analgesic Adjunct to General Anesthesia | Adults | IV | Low Dose: 1-2 mcg/kg. Moderate Dose: 2-8 mcg/kg. | Incremental: 10-25 mcg as needed. Infusion: Adjust to maintain analgesia, not to exceed 1 mcg/kg/hr total dose. | Used with other anesthetics. Lower doses for shorter, less stimulating procedures. 20 |
Primary Anesthetic Agent | Adults | IV | 8-30 mcg/kg | Incremental: 25-50 mcg as needed. Infusion: Titrated to patient response and surgical stimuli. | Administered with 100% oxygen and a neuromuscular blocker. For major surgeries (e.g., cardiac, neurosurgery). 5 |
Anesthesia | Pediatrics (2-12 yrs) | IV | 10-25 mcg/kg (as part of induction) | Incremental doses as needed. | Safety and efficacy not established in children <2 years. Dosing is highly specialized. 26 |
Epidural Analgesia (Labor & Delivery) | Parturients | Epidural | 10-15 mcg | Subsequent doses of 10-15 mcg can be given at intervals of not less than 1 hour. | Administered with a low-dose local anesthetic (e.g., 10 mL of 0.125% bupivacaine). 5 |
Acute Pain Management (Dsuvia®) | Adults | Sublingual | 30 mcg | 30 mcg as needed, with a minimum of 1 hour between doses. Maximum 12 tablets (360 mcg) per 24 hours. | For use only in certified, medically supervised settings. Not for home use. Maximum duration 72 hours. 24 |
Table 4.1: Recommended Dosing Guidelines for Sufentanil by Indication and Route | | | | | |
Section 5: Comparative Analysis: Sufentanil, Fentanyl, and Morphine
To fully appreciate the clinical role of sufentanil, it is essential to compare it with its parent compound, fentanyl, and the archetypal opioid, morphine. This comparison highlights the unique combination of properties that defines sufentanil's specific niche in anesthesia and analgesia.
5.1 Potency and Receptor Affinity
The most striking difference among these three opioids is their relative potency, which is a function of their affinity and efficacy at the μ-opioid receptor.
- Potency Hierarchy: Sufentanil is the most potent of the three. It is estimated to be approximately 5 to 10 times more potent than fentanyl and 500 to 1,000 times more potent than morphine on a weight basis.[1] This is reflected in equianalgesic dosing charts, where an intravenous dose of 10-40 mcg of sufentanil is considered roughly equivalent to 100 mcg of fentanyl or 10,000 mcg (10 mg) of morphine.[29]
- Receptor Selectivity: Sufentanil exhibits a higher degree of selectivity for the μ-opioid receptor compared to fentanyl and morphine.[4] This enhanced selectivity may contribute to its remarkably high therapeutic index, as it may produce fewer off-target effects at therapeutic concentrations compared to less selective opioids.
5.2 Onset and Duration of Action
The speed of onset and duration of effect are critical determinants of an opioid's clinical utility, and these are largely governed by physicochemical properties like lipophilicity.
- Onset of Action: Both sufentanil and fentanyl are highly lipophilic, allowing them to rapidly cross the blood-brain barrier and produce a fast onset of action. The time to effect-site equilibration (t1/2ke0) is similar for both, at approximately 6.2 minutes for sufentanil and 6.6 minutes for fentanyl.[4] In contrast, morphine is significantly more hydrophilic (less lipophilic). This property hinders its passage into the CNS, resulting in a much slower onset of central effects, with a
t1/2ke0 of approximately 3 hours.[4]
- Duration and Recovery: The duration of action is more complex and is best understood by considering the context-sensitive half-time (CSt1/2), which describes the time for the plasma concentration to decrease by 50% after stopping a continuous infusion of a specific duration.
- Sufentanil vs. Fentanyl: After a single bolus, both have a short duration due to rapid redistribution. For continuous infusions of moderate duration (up to approximately 6-8 hours), sufentanil generally has a shorter CSt1/2 than fentanyl, which may allow for a more rapid recovery and extubation.[4] For very prolonged infusions (>8 hours), this advantage may diminish or reverse, with alfentanil potentially offering the fastest recovery.[31]
- Morphine: Morphine has the longest duration of action, which is advantageous for providing sustained postoperative analgesia after a single neuraxial dose but also carries a significant risk of delayed respiratory depression, as plasma concentrations decline much more slowly and it has active metabolites.[30]
5.3 Clinical Efficacy and Therapeutic Index
- Therapeutic Index (TI): The TI, a measure of the margin of safety between the therapeutic and toxic doses, is exceptionally high for sufentanil. In preclinical models, its TI is estimated to be around 26,700, compared to approximately 280 for fentanyl and 70 for morphine.[4] This suggests that in controlled experimental conditions, there is a very wide margin between the dose required for analgesia and the dose causing severe toxicity.
- Clinical Efficacy: All three are highly effective analgesics. The choice depends on the clinical goal. For blunting the intense but brief stress of laryngoscopy, a bolus of sufentanil is five times more potent than fentanyl.[4] For prolonged postoperative analgesia from a single intrathecal injection, morphine is superior to fentanyl due to its longer duration.[30] Sufentanil's primary advantage in efficacy lies in its profound attenuation of the sympathetic stress response during major surgery, providing superior hemodynamic stability.[4]
5.4 Comparative Side-Effect Profiles
While all three agents share the classic opioid class side effects (e.g., respiratory depression, sedation, nausea, vomiting, constipation, pruritus), there are notable differences in their profiles.
- Sufentanil: Is particularly associated with producing significant bradycardia and has a notable risk of skeletal muscle rigidity, especially with rapid, high-dose IV injection.[5]
- Fentanyl: Is generally considered to induce less histamine release than morphine, resulting in less itching and hypotension.[33]
- Morphine: Is the most likely of the three to cause histamine release from mast cells, which can lead to significant hypotension, flushing, and bronchoconstriction in susceptible individuals.[4] Its long half-life and active metabolite (morphine-6-glucuronide) contribute to a higher risk of delayed and prolonged respiratory depression, especially with repeated dosing or in patients with renal impairment.[30]
The clinical positioning of sufentanil is not merely as a "stronger fentanyl" but is defined by a nuanced trade-off between its extreme potency and its controllability. While its potency allows for the use of very small volumes and the achievement of profound anesthetic depth and hemodynamic stability, it also means that the margin for a dosing error is exceedingly small. The consequences of an overdose, particularly profound and rapid-onset respiratory depression, are immediate and severe. Although its pharmacokinetic profile, specifically its shorter context-sensitive half-time after moderate-length infusions, can offer faster recovery times compared to fentanyl, its terminal elimination half-life remains significant, carrying a persistent risk of renarcotization if monitoring is not vigilant. Therefore, the decision to use sufentanil over the more familiar fentanyl is a calculated clinical judgment. The practitioner is trading the wider margin for error and predictability of fentanyl for the specific benefits of sufentanil's superior hemodynamic control and potentially faster recovery. This is a trade-off that is only appropriate for highly skilled specialists operating in tightly controlled environments, such as cardiac operating rooms, where the specific benefits of sufentanil are most needed and its heightened risks can be expertly managed.
Characteristic | Morphine | Fentanyl | Sufentanil | Source(s) |
---|
Relative Potency (IV, vs. Morphine=1) | 1 | 50-100x | 500-1,000x | 1 |
Receptor Selectivity | μ-agonist | μ-agonist | Highly selective μ-agonist | 4 |
Lipophilicity | Low (Hydrophilic) | High | Very High | 4 |
Onset of Action (IV, CNS Effect) | Slow (~3 hours to peak effect) | Rapid (~6.6 min to peak effect) | Rapid (~6.2 min to peak effect) | 4 |
CSt1/2 (4h infusion) | Long | ~250 min | ~30 min | 4 |
Terminal Half-life | 2-3 hours (active metabolites) | 3-4 hours | ~2.7 hours | 1 |
Key Side Effects | Histamine release, delayed respiratory depression | Muscle rigidity, dizziness | Profound bradycardia, muscle rigidity | 4 |
Primary Clinical Niche | Post-op pain (PCA), long-acting neuraxial analgesia | General anesthesia, chronic pain (patch), procedural sedation | Anesthesia for major surgery (cardiac), labor epidurals | 3 |
Table 5.1: Comparative Pharmacological and Clinical Profile of Sufentanil, Fentanyl, and Morphine | | | | |
Section 6: Safety, Tolerability, and Risk Management
The immense potency of sufentanil necessitates a comprehensive understanding of its safety profile and the implementation of rigorous risk management strategies. This section details its adverse effects, contraindications, overdose management, and the specific regulatory controls applied to its sublingual formulation.
6.1 Adverse Drug Reactions
The adverse effects of sufentanil are predominantly extensions of its opioid agonist activity. They are dose-dependent and can affect multiple organ systems.
- Most Frequent Adverse Reactions: Based on clinical trial data, the most commonly reported adverse effects (≥1/10 frequency) are nausea and vomiting. Other common reactions include headache, dizziness, hypotension, and pyrexia (fever).[24]
- Most Serious Adverse Reaction: The most critical and life-threatening adverse reaction is respiratory depression. This can range in severity from a reduced respiratory rate (bradypnea) and hypoventilation to complete apnea and respiratory arrest. This risk is inherent to all opioids but is particularly acute with sufentanil due to its potency and rapid onset.[10]
- Other Clinically Significant Reactions:
- Cardiovascular: Bradycardia, hypotension (including orthostatic hypotension), and syncope are common.[3]
- Neurological: Sedation, somnolence, confusion, and dizziness are frequent. High doses or rapid injection can cause severe skeletal muscle rigidity, particularly of the chest wall, which can interfere with ventilation.[3]
- Gastrointestinal: Constipation due to reduced GI motility is a very common effect with sustained use. Spasm of the sphincter of Oddi can also occur.[3]
- Dermatological: Pruritus (itching) is common, particularly with neuraxial (epidural or intrathecal) administration.[3]
6.2 U.S. Boxed Warnings and Major Precautions
The U.S. FDA requires boxed warnings for medications with serious or life-threatening risks. Sufentanil's labeling includes several critical warnings that underscore the need for extreme caution.[10]
- Addiction, Abuse, and Misuse: As a potent opioid, sufentanil exposes users to the significant risks of opioid use disorder (OUD), addiction, abuse, and misuse, which can lead to overdose and death. A patient's risk should be assessed prior to administration and monitored throughout treatment.
- Life-Threatening Respiratory Depression: Serious, life-threatening, or fatal respiratory depression can occur, even at recommended doses. The risk is highest during the initiation of therapy or following a dosage increase.
- Accidental Exposure (Sublingual Tablet): Accidental ingestion of even one sublingual tablet, especially by a child or an opioid-naïve individual, can result in a fatal overdose. This specific risk is the primary driver for the Dsuvia® REMS program.
- Cytochrome P450 3A4 Interaction: The concomitant use of sufentanil with CYP3A4 inhibitors can significantly increase sufentanil plasma concentrations, potentially leading to a fatal overdose. Conversely, discontinuing a CYP3A4 inducer can have the same effect.
- Risks from Concomitant Use with Benzodiazepines or Other CNS Depressants: Co-administration with other CNS depressants, including benzodiazepines and alcohol, poses a profound risk of additive effects, leading to deep sedation, respiratory depression, coma, and death. This combination should be reserved for patients for whom alternative options are inadequate.
6.3 Contraindications
Sufentanil is strictly contraindicated in certain clinical situations where its risks are likely to outweigh any potential benefits.
- Patients with significant respiratory depression.[24]
- Patients with acute or severe bronchial asthma in an unmonitored setting or in the absence of resuscitative equipment.[24]
- Patients with known or suspected gastrointestinal obstruction, including paralytic ileus.[10]
- Patients with a known hypersensitivity to sufentanil or any of its components.[36]
6.4 Overdose: Manifestations and Management
An overdose of sufentanil is a medical emergency that requires immediate intervention.
- Manifestations: The clinical presentation of an overdose is an exaggeration of its pharmacological effects. Key signs include severe respiratory depression, somnolence progressing to stupor or coma, skeletal muscle flaccidity, cold and clammy skin, and constricted pupils (miosis). In the presence of severe hypoxia, pupils may become dilated (mydriasis), which can be a misleading sign.[20]
- Management:
- Airway and Ventilation: The immediate priority is the re-establishment of a patent airway and the institution of assisted or controlled ventilation with oxygen.
- Antidote Administration: The specific antidote for opioid-induced respiratory depression is an opioid antagonist, such as naloxone. Naloxone acts as a competitive antagonist at the μ-opioid receptor, displacing sufentanil and reversing its effects.[1]
- Vigilant Monitoring: A critical aspect of management is recognizing that the duration of action of naloxone (typically 30-90 minutes) is often shorter than the duration of action of sufentanil, especially after a large dose or with sustained-release formulations. This creates a risk of "renarcotization," where the patient's respiratory depression returns as the naloxone wears off. Therefore, continuous monitoring and repeated doses or a continuous infusion of naloxone may be necessary until the patient's spontaneous respiration is reliably re-established.[1]
- Supportive Care: Other supportive measures, such as vasopressors for circulatory shock and management of pulmonary edema, should be employed as indicated.[20]
6.5 The Dsuvia® (Sufentanil Sublingual Tablet) REMS Program
The FDA mandated a Risk Evaluation and Mitigation Strategy (REMS) program for the Dsuvia® sublingual tablet formulation due to its unique and significant risk profile. This program is a clear example of formulation-driven risk management. While the active drug, sufentanil, has been used for decades in liquid form (Sufenta®) without a REMS, its use was inherently confined to the highly controlled, sterile "closed-loop" environment of operating rooms and labor wards, handled almost exclusively by anesthesiologists. The creation of a solid, single-dose sublingual tablet, while innovative for its ease of administration, introduced a new and substantial risk vector. This portable solid form is far more susceptible to being dropped, lost, diverted, or accidentally ingested than a vial of liquid. The potential for a child or opioid-naïve adult to find and consume a single 30 mcg tablet—a potentially lethal dose—is vastly higher. The Dsuvia® REMS program was designed specifically to counteract this new risk. Its core requirements—prohibiting retail pharmacy access, restricting use to certified inpatient settings, and mandating staff training—effectively recreate the "closed-loop" control of the operating room for this new, more portable formulation, preventing the drug from leaving a supervised environment.
- Rationale: The REMS was instituted to mitigate the risk of life-threatening respiratory depression resulting from accidental exposure to the highly potent sublingual tablet.[13]
- Core Requirements:
- Restricted Distribution and Dispensing: Dsuvia® is not available in retail pharmacies and is strictly prohibited from being dispensed for outpatient or home use.[14] Treatment must be discontinued before the patient is discharged.[27]
- Healthcare Setting Certification: Only medically supervised healthcare settings (e.g., hospitals, surgical centers, emergency departments) that have the capacity to manage acute opioid overdose are eligible to dispense and administer Dsuvia®. To become certified, the facility must enroll in the REMS program, designate an authorized representative to oversee compliance, and establish clear processes and procedures.[14]
- Staff Training: All relevant staff involved in the administration or handling of Dsuvia® must be trained on the REMS requirements. This includes understanding the prohibition on outpatient dispensing and the necessity of referring to the Directions for Use before administration.[14]
- Audits and Compliance: Certified healthcare settings must maintain records of training and procedures and must agree to comply with audits conducted by the manufacturer to ensure that all elements of the REMS program are being followed.[14]
Section 7: Significant Drug-Drug Interactions
The safe use of sufentanil requires careful consideration of potential drug-drug interactions, which can be broadly categorized by their underlying mechanism as either pharmacodynamic (affecting the drug's action at its receptor) or pharmacokinetic (affecting the drug's ADME profile).
7.1 Interactions with CNS Depressants (Pharmacodynamic Synergism)
- Interacting Drugs: This class includes benzodiazepines, alcohol, other opioids (e.g., hydrocodone), general anesthetics (e.g., propofol), phenothiazines, and skeletal muscle relaxants.[10]
- Mechanism and Outcome: Concomitant use of sufentanil with any other CNS depressant results in additive or synergistic pharmacodynamic effects. Both drugs suppress CNS function, leading to a dangerously amplified risk of profound sedation, life-threatening respiratory depression, severe hypotension, coma, and death. The FDA boxed warning highlights this specific risk, especially with benzodiazepines.[10]
- Management: This combination should be avoided or reserved only for patients in whom alternative treatment options are inadequate. If co-administration is necessary, dosages and durations should be limited to the minimum required, and patients must be monitored closely for signs of respiratory depression and sedation.[10]
7.2 Cytochrome P450 3A4 Inhibitors and Inducers (Pharmacokinetic Interaction)
- Interacting Drugs:
- CYP3A4 Inhibitors: This group includes potent inhibitors such as azole antifungals (e.g., ketoconazole, itraconazole), protease inhibitors (e.g., ritonavir), many macrolide antibiotics (e.g., clarithromycin, erythromycin), and grapefruit juice.[21]
- CYP3A4 Inducers: This group includes drugs like rifampin, carbamazepine, and phenytoin.[34]
- Mechanism and Outcome: Sufentanil is primarily metabolized by the CYP3A4 enzyme system.
- Inhibitors: Co-administration with a CYP3A4 inhibitor blocks the metabolic breakdown of sufentanil. This leads to elevated and prolonged plasma concentrations of sufentanil from a standard dose, which can cause increased or prolonged adverse effects, including a potentially fatal overdose from respiratory depression. A clinical study demonstrated that the potent inhibitor ketoconazole increased the total exposure (AUC) to sublingual sufentanil by 77%.[21]
- Inducers: Co-administration with a CYP3A4 inducer accelerates the metabolism of sufentanil. This can lead to decreased plasma concentrations and reduced analgesic efficacy. In patients with physical dependence, this may precipitate opioid withdrawal symptoms.[34]
- Management: Patients receiving sufentanil concomitantly with a CYP3A4 inhibitor or after discontinuing a CYP3A4 inducer must be monitored closely for signs of overdose. Dose reduction of sufentanil may be necessary. Conversely, patients starting a CYP3A4 inducer or stopping an inhibitor may require an increased sufentanil dose and should be monitored for signs of reduced efficacy or withdrawal.[10]
7.3 Serotonergic Agents and Serotonin Syndrome
- Interacting Drugs: This includes various classes of antidepressants such as Selective Serotonin Reuptake Inhibitors (SSRIs), Serotonin-Norepinephrine Reuptake Inhibitors (SNRIs), Tricyclic Antidepressants (TCAs), and Monoamine Oxidase Inhibitors (MAOIs), as well as triptans and other drugs that affect serotonin levels.[10]
- Mechanism and Outcome: Opioids, including sufentanil, have serotonergic activity. When combined with other serotonergic agents, there is an increased risk of developing serotonin syndrome, a potentially life-threatening condition characterized by a triad of autonomic hyperactivity, neuromuscular abnormalities, and altered mental status. Symptoms can include agitation, hallucinations, tachycardia, fever, muscle rigidity, and seizures. The risk is particularly high with MAOIs.[37]
- Management: Patients should be monitored for symptoms of serotonin syndrome. If suspected, sufentanil and the concomitant serotonergic agent should be discontinued immediately.[20] Use with MAOIs is generally not recommended or is contraindicated.[39]
7.4 Mixed Agonist/Antagonist and Partial Agonist Opioids
- Interacting Drugs: This class includes buprenorphine (a partial μ-agonist), nalbuphine, butorphanol, and pentazocine (mixed agonist-antagonists).[20]
- Mechanism and Outcome: These drugs can bind to the μ-opioid receptor and act as competitive antagonists or partial agonists in the presence of a full agonist like sufentanil. This interaction can reduce the analgesic effect of sufentanil. More importantly, in patients who are physically dependent on a full opioid agonist, administration of these drugs can precipitate an acute and severe withdrawal syndrome.[20]
- Management: The use of mixed agonist/antagonist or partial agonist opioids should be avoided in patients receiving sufentanil.[20]
Interacting Drug Class | Example Drugs | Mechanism of Interaction | Clinical Consequence | Recommended Management |
---|
CNS Depressants | Benzodiazepines, Alcohol, Propofol | Pharmacodynamic Synergism | Profound sedation, respiratory depression, hypotension, coma, death. | Avoid combination or limit dose/duration; monitor closely. 10 |
CYP3A4 Inhibitors | Ketoconazole, Ritonavir, Clarithromycin | Pharmacokinetic (Inhibition of Metabolism) | Increased sufentanil plasma concentration, risk of fatal overdose. | Monitor closely for toxicity; consider sufentanil dose reduction. 10 |
CYP3A4 Inducers | Rifampin, Carbamazepine, Phenytoin | Pharmacokinetic (Induction of Metabolism) | Decreased sufentanil plasma concentration, reduced efficacy, risk of withdrawal. | Monitor for efficacy; consider sufentanil dose increase. 34 |
Serotonergic Agents | SSRIs, SNRIs, MAOIs, Triptans | Pharmacodynamic (Excess Serotonergic Activity) | Increased risk of life-threatening Serotonin Syndrome. | Monitor for symptoms; discontinue if suspected. Avoid use with MAOIs. 10 |
Mixed/Partial Agonist Opioids | Buprenorphine, Nalbuphine, Pentazocine | Pharmacodynamic (Receptor Competition) | Reduced analgesia and/or precipitation of withdrawal symptoms. | Avoid concomitant use. 20 |
Table 7.1: Clinically Significant Drug Interactions with Sufentanil | | | | |
Section 8: Considerations in Special Populations
The safe and effective use of sufentanil is highly dependent on patient-specific factors. Its pharmacokinetics and pharmacodynamics can be significantly altered in certain populations, necessitating careful dose adjustments and heightened monitoring.
8.1 Pediatric and Geriatric Use
The risk profile of sufentanil is not linear across the lifespan but rather follows a U-shaped curve, with the greatest risks observed at the extremes of life—in neonates and the elderly. This pattern is driven by age-related changes in pharmacokinetics and pharmacodynamics. Neonates are at high pharmacokinetic risk due to their immature hepatic metabolism, lower levels of the binding protein AAG (which increases the free, active drug fraction), and consequently, reduced clearance and a prolonged elimination half-life. In contrast, children between the ages of 2 and 12 exhibit faster clearance and a shorter half-life than adults, altering their dosing requirements. As individuals enter older age, the risk rises again. Geriatric patients often have reduced hepatic and renal function, decreased physiological reserve, and a higher prevalence of comorbidities and polypharmacy. These factors increase both pharmacokinetic and pharmacodynamic risks, making them more sensitive to the drug's effects, particularly respiratory depression and hypotension. This U-shaped risk profile mandates distinct, age-specific clinical approaches to dosing and monitoring.
- Pediatric Population:
- Pharmacokinetics: The disposition of sufentanil is highly variable in children. Neonates exhibit a markedly prolonged elimination half-life (mean ~434 minutes) and reduced clearance compared to adults, placing them at a very high risk of drug accumulation and toxicity.[3] Conversely, children aged 2 to 8 years have been shown to have a shorter half-life (mean ~97 minutes) and a faster clearance rate than adults, which may necessitate different dosing strategies to maintain therapeutic effect.[5]
- Clinical Use: For general anesthesia, safety and efficacy have not been established in children under 2 years of age.[26] Dosing in older children (2-12 years) must be highly specialized and carefully titrated.[26] The sublingual tablet formulation (Dsuvia®) is explicitly not approved for use in any pediatric population.[14]
- Geriatric Population:
- Increased Sensitivity: Elderly patients are generally more sensitive to the therapeutic and adverse effects of opioids. They may experience more profound sedation, confusion, and respiratory depression from a given dose.
- Pharmacokinetics: Age-related declines in hepatic and renal function can lead to decreased clearance and a prolonged duration of action.
- Management: Dosing in geriatric patients should be initiated at the lower end of the recommended range, and titration should proceed cautiously. Close monitoring for adverse effects, particularly respiratory depression, CNS depression, and hypotension, is essential.[26]
8.2 Use in Pregnancy and Lactation
The use of sufentanil during pregnancy and lactation requires a careful balancing of maternal benefit against potential fetal and neonatal risk.
- Pregnancy: Sufentanil is known to cross the placental barrier.[37] Its use during labor and delivery can cause respiratory depression in the newborn. Prolonged maternal use during pregnancy can lead to neonatal opioid withdrawal syndrome, characterized by irritability, high-pitched crying, tremors, and feeding difficulties. Therefore, sufentanil is not recommended for use during pregnancy unless the potential benefit to the mother is judged to outweigh the potential risk to the fetus. If used near term, an antidote for the infant (naloxone) should be immediately available.[22]
- Lactation: Sufentanil is excreted into human milk.[22] Although amounts ingested by the infant after short-term maternal use (e.g., epidural during labor) are likely small, there is a potential for adverse effects, especially with repeated or prolonged maternal use. Breastfed infants of mothers receiving sufentanil should be closely monitored for signs of excess sedation, difficulty feeding, and respiratory depression. A decision must be made whether to discontinue breastfeeding or discontinue the drug, taking into account the importance of the drug to the mother.[3]
8.3 Patients with Hepatic or Renal Impairment
Since sufentanil is metabolized by the liver and its metabolites are excreted by the kidneys, organ impairment can significantly alter its pharmacokinetic profile.
- Hepatic Impairment: As sufentanil is extensively metabolized in the liver, its clearance may be substantially reduced in patients with moderate to severe hepatic impairment. This can lead to higher plasma concentrations and a prolonged duration of effect, increasing the risk of toxicity. Sufentanil should be administered with caution in this population, and patients should be monitored carefully for signs of overdose.[36]
- Renal Impairment: While only a small amount of sufentanil is excreted unchanged, its inactive metabolites are cleared by the kidneys. In patients with severe renal impairment, the elimination of these metabolites may be altered. Although the clinical significance is less clear than with hepatic impairment, caution and close monitoring for adverse effects are recommended when administering sufentanil to patients with severe renal disease.[36] A clinical trial has been completed to investigate the use of sufentanil in patients with liver dysfunction, which may provide more specific guidance in the future.[44]
Section 9: Expert Synthesis and Concluding Remarks
Sufentanil stands as a therapeutic agent of profound duality. Its chemical architecture confers an unparalleled level of potency and a unique set of pharmacological properties that, in the right context, are of immense clinical value. The combination of rapid onset, profound analgesia, and a remarkable ability to maintain hemodynamic stability under extreme surgical stress makes it an indispensable tool for anesthesiologists managing high-risk patients in settings such as cardiac and major neurosurgery.[3] In these specific scenarios, it offers clinical advantages in controlling the patient's physiological response to surgery that are difficult, if not impossible, to replicate with other available agents.
Conversely, the very properties that make sufentanil so valuable also render it one of the most hazardous opioids if used improperly or outside of a strictly controlled environment. Its extreme potency results in a narrow therapeutic window in clinical practice, where even minor miscalculations in dosage can lead to immediate and severe consequences, most notably life-threatening respiratory depression. The significant liabilities associated with error, accidental exposure, or diversion demand a level of vigilance and systemic control that exceeds that of most other medications.
The evolution of sufentanil's formulations provides a compelling case study of the central challenge in modern pain management: the continuous drive for more effective, faster-acting, and more convenient analgesic options must be perpetually balanced against the societal imperative of ensuring public safety and preventing the diversion and misuse that fuel the opioid crisis. The transition from the specialist-only intravenous solution (Sufenta®), with its inherently limited access, to the innovative sublingual tablet (Dsuvia®), designed for broader use within acute care settings, exemplifies this tension. The latter formulation, while offering a non-invasive and rapid method of pain relief, simultaneously created a new risk of accidental exposure that necessitated the creation of a stringent, closed-system regulatory framework in the form of the REMS program.
Ultimately, the safe and effective application of sufentanil is less a function of the drug molecule itself and more a testament to the critical importance of the system in which it is deployed. The benefit-risk profile of this powerful agent is only favorable when it is administered by highly trained and experienced professionals, within certified healthcare settings that are equipped to manage its consequences, and according to robust, evidence-based protocols for dosing, continuous monitoring, and immediate overdose management. The Dsuvia® REMS program is a clear embodiment of this principle. The future of sufentanil and other potent opioids in medicine will be defined not by their pharmacological potential alone, but by our collective ability to design, implement, and adhere to these kinds of comprehensive, fail-safe systems of care that protect both the patient and the public.
Works cited
- [Sufentanil - Wikipedia, accessed July 24, 2025, ][https://en.wikipedia.org/wiki/Sufentanil]
- [Sufentanil: Pharmacolgy and Current Applications in Clinical Practice - JSciMed Central, accessed July 24, 2025, ][https://www.jscimedcentral.com/jounal\-article\-info/Journal\-of\-Pharmacology\-and\-Clinical\-Toxicology/Sufentanil:\-Pharmacolgy\-and\-Current\-Applications\-in\-Clinical\-Practice\-10739]
- [Sufentanil: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed July 24, 2025, ][https://go.drugbank.com/drugs/DB00708]
- [Sufentanil: Pharmacolgy and Current Applications ... - JSciMed Central, accessed July 24, 2025, ][https://www.jscimedcentral.com/public/assets/articles/pharmacology\-10\-1165\.pdf]
- [Sufentanil Citrate Injection, USP CII - accessdata.fda.gov, accessed July 24, 2025, ][https://www.accessdata.fda.gov/drugsatfda\_docs/label/2014/019050s032lbl.pdf]
- [Drug ID - ADReCS, accessed July 24, 2025, ][https://www.bio\-add.org/ADReCS/drugSummary.jsp?drug\_id\=BADD\_D02082\&uname\=8348a7f3\-b72a\-4a05\-bb6d\-7bfa6fb69d02]
- [Epidural sufentanil (Chapter 43) - The Essence of Analgesia and Analgesics, accessed July 24, 2025, ][https://www.cambridge.org/core/books/essence\-of\-analgesia\-and\-analgesics/epidural\-sufentanil/C1FF0257B87757903F29D641B8F5FEA3]
- [Sufentanil | C22H30N2O2S | CID 41693 - PubChem, accessed July 24, 2025, ][https://pubchem.ncbi.nlm.nih.gov/compound/Sufentanil]
- [Sublingual sufentanil tablet for analgesia in emergency medical services and search and rescue agenc - Handtevy, accessed July 24, 2025, ][https://www.handtevy.com/wp\-content/uploads/2024/11/Sublingual\-sufentanil\-tablet\-for\-analgesia\-in\-emergency\-medical\-services\-and\-search\-and\-rescue\-agencies.pdf]
- [SUFentanil | Drug Lookup | Pediatric Care Online - AAP Publications, accessed July 24, 2025, ][https://publications.aap.org/pediatriccare/drug\-monograph/18/5566/SUFentanil]
- [Reference ID: 4027921 - Amazon S3, accessed July 24, 2025, ][https://s3\-us\-west\-2\.amazonaws.com/drugbank/fda\_labels/DB00708\.pdf?1541440044]
- [SUFENTANIL CITRATE injection, solution - DailyMed, accessed July 24, 2025, ][https://dailymed.nlm.nih.gov/dailymed/drugInfo.cfm?setid\=20293943\-46ff\-4345\-1aa4\-929b4e017a25]
- [What is the Dsuvia REMS program? - Drugs.com, accessed July 24, 2025, ][https://www.drugs.com/medical\-answers/dsuvia\-rems\-program\-3457015/]
- [REMS Information for DSUVIA | Home, accessed July 24, 2025, ][https://www.dsuviarems.com/]
- [Global research on sufentanil use in anesthesiology from 2003 to 2023: a bibliometric analysis - Frontiers, accessed July 24, 2025, ][https://www.frontiersin.org/journals/pharmacology/articles/10\.3389/fphar.2024\.1412726/full]
- [sufentanil | Ligand page - IUPHAR/BPS Guide to PHARMACOLOGY, accessed July 24, 2025, ][https://www.guidetopharmacology.org/GRAC/LigandDisplayForward?ligandId\=3534]
- [Sufentanil (R 30730, CAS Number: 56030-54-7) | Cayman Chemical, accessed July 24, 2025, ][https://www.caymanchem.com/product/42344/sufentanil]
- [Sufentanil Base | CAS 56030-54-7 - Veranova, accessed July 24, 2025, ][https://veranova.com/controlled\-substances/sufentanil\-base/]
- [Formulary Drug Review: Sufentanil Sublingual - PMC - PubMed Central, accessed July 24, 2025, ][https://pmc.ncbi.nlm.nih.gov/articles/PMC6628549/]
- [SUFENTANIL CITRATE INJECTION - accessdata.fda.gov, accessed July 24, 2025, ][https://www.accessdata.fda.gov/drugsatfda\_docs/label/2023/019050s043lbl.pdf]
- [Pharmacokinetic Properties of a Sufentanil Sublingual Tablet Intended to Treat Acute Pain - PubMed, accessed July 24, 2025, ][https://pubmed.ncbi.nlm.nih.gov/29498947/]
- [Sufentanil - Drugs and Lactation Database (LactMed®) - NCBI Bookshelf, accessed July 24, 2025, ][https://www.ncbi.nlm.nih.gov/books/NBK501254/]
- [The pharmacokinetics of sufentanil in surgical patients - PubMed, accessed July 24, 2025, ][https://pubmed.ncbi.nlm.nih.gov/6238552/]
- [Sufentanil Sublingual Tablet (Dsuvia) - Medical Clinical Policy Bulletins - Aetna, accessed July 24, 2025, ][https://www.aetna.com/cpb/medical/data/900\_999/0944\.html]
- [sufentanil citrate injection, USP Dosage and Administration | Pfizer Medical - US, accessed July 24, 2025, ][https://www.pfizermedical.com/sufentanil\-citrate/dosage\-admin]
- [Sufenta (sufentanil) dosing, indications, interactions, adverse effects, and more, accessed July 24, 2025, ][https://reference.medscape.com/drug/sufenta\-sufentanil\-343325]
- [Dsuvia Advanced Patient Information - Drugs.com, accessed July 24, 2025, ][https://www.drugs.com/cons/dsuvia.html]
- [Comparison of Effects: Sufentanil vs. Fentanyl | Futures, accessed July 24, 2025, ][https://futuresrecoveryhealthcare.com/blog/sufentanil\-vs\-fentanyl/]
- [Appendix A: Equianalgesic Conversion for Morphine - Gov.bc.ca, accessed July 24, 2025, ][https://www2\.gov.bc.ca/assets/gov/health/practitioner\-pro/bc\-guidelines/palliative2\_\-\_pain\_equianalgesic.pdf]
- [Comparison of the Efficacy and Safety of Morphine and Fentanyl as ..., accessed July 24, 2025, ][https://pmc.ncbi.nlm.nih.gov/articles/PMC5735488/]
- [Rational Opioid Selection - Medic's Corner, accessed July 24, 2025, ][http://www.jvsmedicscorner.com/Anaesth\-Pharmacology\_files/Pharmacokinetics,%20Pharmacodynamics,%20and%20Rational%20opioid%20selection.10\.pdf]
- [Pharmacokinetics, pharmacodynamics, and rational opioid selection - PubMed, accessed July 24, 2025, ][https://pubmed.ncbi.nlm.nih.gov/1824743/]
- [Fentanyl - Wikipedia, accessed July 24, 2025, ][https://en.wikipedia.org/wiki/Fentanyl]
- [Fentanyl - StatPearls - NCBI Bookshelf, accessed July 24, 2025, ][https://www.ncbi.nlm.nih.gov/books/NBK459275/]
- [Fentanyl vs Morphine: Similarities, Differences & Overdose Potential Comparison, accessed July 24, 2025, ][https://www.therecoveryvillage.com/fentanyl\-addiction/fentanyl\-vs\-morphine/]
- [Medicinal Product no longer authorised - EMA, accessed July 24, 2025, ][https://www.ema.europa.eu/en/documents/product\-information/zalviso\-epar\-product\-information\_en.pdf]
- [Dzuveo, INN-sufentanil citrate - EMA, accessed July 24, 2025, ][https://www.ema.europa.eu/en/documents/product\-information/dzuveo\-epar\-product\-information\_en.pdf]
- [Dsuvia - FEPBlue, accessed July 24, 2025, ][https://www.fepblue.org/\-/media/PDFs/Medical\-Policies/2024/March/Mar\-2024\-Pharmacy\-Policies/Remove\-\-\-Replace/570073\-Dsuvia\-sufentanil.pdf]
- [Sufentanil Sublingual Tablets - Cleveland Clinic, accessed July 24, 2025, ][https://my.clevelandclinic.org/health/drugs/21246\-sufentanil\-sublingual\-tablets]
- [Sufentanil (injection route) - Side effects & uses - Mayo Clinic, accessed July 24, 2025, ][https://www.mayoclinic.org/drugs\-supplements/sufentanil\-injection\-route/description/drg\-20406181]
- [publications.aap.org, accessed July 24, 2025, ][https://publications.aap.org/pediatriccare/drug\-monograph/18/5566/SUFentanil\#:\~:text\=The%20concomitant%20use%20of%20sufentanil,cause%20potentially%20fatal%20respiratory%20depression.]
- [Sufentanil - Memorial Sloan Kettering Cancer Center, accessed July 24, 2025, ][https://www.mskcc.org/cancer\-care/patient\-education/medications/adult/sufentanil]
- [Dsuvia (sufentanil SL) dosing, indications, interactions, adverse effects, and more, accessed July 24, 2025, ][https://reference.medscape.com/drug/dsuvia\-sufentanil\-sl\-999947]
- [Liver Dysfunction Completed Phase Trials for Sufentanil (DB00708) | DrugBank Online, accessed July 24, 2025, ][https://go.drugbank.com/indications/DBCOND0029904/clinical\_trials/DB00708?phase\=\&status\=completed]