C22H30N2O2S
1401028-24-7
Severe Acute Pain
Oliceridine is a novel, synthetic, intravenously administered opioid agonist approved by the U.S. Food and Drug Administration (FDA) for the management of acute pain in adults that is severe enough to require an intravenous opioid and for whom alternative treatments are inadequate.[1] Marketed under the brand name Olinvyk, it is classified as a Schedule II controlled substance, reflecting its potential for abuse and dependence.[1] Oliceridine represents the first-in-class G protein-biased agonist at the μ-opioid receptor (MOR). This mechanism is designed to preferentially activate the G protein-coupled signaling pathway, which is primarily responsible for analgesia, while minimizing the recruitment and activation of the β-arrestin 2 pathway, a cascade implicated in many of the hallmark adverse effects of conventional opioids, including respiratory depression and gastrointestinal dysfunction.[4]
The clinical development program for oliceridine demonstrated analgesic efficacy that was superior to placebo and non-inferior to morphine in postoperative pain models following both hard tissue (bunionectomy) and soft tissue (abdominoplasty) surgeries.[7] The most pronounced clinical advantage observed in these pivotal trials was a statistically significant improvement in gastrointestinal tolerability, with patients receiving oliceridine experiencing lower rates of nausea and vomiting and requiring less rescue antiemetic medication compared to those treated with morphine at equianalgesic doses.[9]
However, the anticipated clear and significant advantage in respiratory safety was not definitively established in the pre-specified analyses of the Phase III registration trials; the difference between oliceridine and morphine on the composite respiratory safety burden endpoint did not achieve statistical significance.[7] Subsequent post-hoc and retrospective analyses suggest a more favorable respiratory profile, though these findings require prospective confirmation.[12] Furthermore, human abuse potential studies indicate that oliceridine possesses a pharmacological profile and abuse liability comparable to equianalgesic doses of morphine.[3]
Reflecting a careful balance of its benefits and risks, particularly a potential for QTc interval prolongation at higher exposures, the use of oliceridine is subject to critical restrictions. It is indicated only for short-term use in controlled clinical settings and is governed by a maximum recommended cumulative daily dose of 27 mg.[14] Oliceridine is thus positioned not as a universal replacement for conventional opioids, but as a specialized therapeutic option within the acute pain armamentarium, offering a distinct risk-benefit profile that may be particularly advantageous for patients at high risk for postoperative nausea and vomiting.
Oliceridine is a small molecule drug developed by Trevena, Inc., and was previously known by the developmental code TRV-130.[16] Upon receiving regulatory approval, it was assigned the brand name Olinvyk, having been referred to as Olinvo during its later stages of development.[18] For precise identification across chemical, regulatory, and biomedical databases, oliceridine is cataloged under numerous unique identifiers, which are consolidated in Table 1.
Table 1: Oliceridine Identifiers and Chemical Properties
Identifier Type | Value | Source Database(s) |
---|---|---|
DrugBank ID | DB14881 | DrugBank 1 |
CAS Number | 1401028-24-7 | PubChem, ChEMBL 1 |
UNII | MCN858TCP0 | FDA GSRS 1 |
DEA Code Number | 9245 | DEA 1 |
PubChem CID | 66553195 | PubChem 14 |
ChEMBL ID | CHEMBL2443262 | ChEMBL 14 |
KEGG ID | D11214 | KEGG 1 |
IUPAC Name | N-[(3-methoxythiophen-2-yl)methyl]-2-decan-9-yl]ethanamine | PubChem 1 |
Oliceridine possesses the molecular formula C22H30N2O2S and a molecular weight of approximately 386.56 g/mol.[1] Its complex chemical structure is unambiguously defined by standardized chemical notations. The International Union of Pure and Applied Chemistry (IUPAC) name is N-[(3-methoxythiophen-2-yl)methyl]-2-decan-9-yl]ethanamine, which describes the specific arrangement of its thiophene, pyridine, and spirocyclic moieties.[1]
For computational and cheminformatic applications, its structure is represented by the following:
Calculated physicochemical properties provide insight into its drug-like characteristics. With a calculated partition coefficient (AlogP) of 4.69 and a topological polar surface area (TPSA) of 43.38 A˚2, oliceridine is a lipophilic molecule capable of crossing biological membranes.[16] It possesses one hydrogen bond donor and five hydrogen bond acceptors (four by Lipinski criteria), seven rotatable bonds, and two aromatic rings.[16] These properties are consistent with Lipinski's Rule of Five, with zero violations reported, indicating favorable oral bioavailability characteristics, although the drug is formulated for intravenous use only.[16] As a base with a predicted basic pKa of approximately 9.12, it exists primarily in its protonated, cationic form at physiological pH.[16]
The pharmacological actions of oliceridine, like all opioid analgesics, are mediated through agonism of the μ-opioid receptor (MOR), a member of the G protein-coupled receptor (GPCR) superfamily.[23] Upon agonist binding, the MOR undergoes a conformational change that triggers two distinct intracellular signaling cascades.
The canonical pathway, responsible for analgesia, involves the coupling and activation of inhibitory G proteins (Gi/o).[23] This activation leads to the dissociation of the G protein into its Gα and Gβγ subunits. The Gα subunit inhibits the enzyme adenylyl cyclase, reducing intracellular levels of cyclic adenosine monophosphate (cAMP). Concurrently, the Gβγ subunit directly modulates ion channels, leading to the opening of G protein-coupled inwardly-rectifying potassium (GIRK) channels and the inhibition of voltage-gated calcium channels. The combined effect of these actions is hyperpolarization of the neuronal membrane and reduced neurotransmitter release, which ultimately dampens the transmission of nociceptive signals in the central nervous system.[5]
A second, distinct pathway is initiated following receptor phosphorylation by GPCR kinases (GRKs). This phosphorylation promotes the binding of an intracellular protein, β-arrestin 2, to the receptor.[23] The recruitment of β-arrestin 2 serves multiple functions: it sterically hinders further G protein coupling, leading to desensitization of the analgesic signal; it facilitates receptor internalization (endocytosis), removing it from the cell surface; and it acts as a scaffold to initiate its own unique signaling cascades.[5] The β-arrestin pathway has been mechanistically linked to many of the dose-limiting adverse effects of conventional opioids, including respiratory depression, constipation, and the development of tolerance.[6]
The central innovation in the design of oliceridine is its property of functional selectivity, also known as "biased agonism".[1] Oliceridine binds to the MOR and preferentially activates the G protein signaling pathway while only minimally recruiting and activating the β-arrestin 2 pathway.[4] In vitro studies demonstrate that oliceridine elicits robust G protein signaling with a potency and efficacy comparable to that of morphine, but with substantially less β-arrestin 2 recruitment and subsequent receptor internalization.[14] This selective activation is hypothesized to uncouple the desired analgesic effects from the adverse effects mediated by β-arrestin, thereby creating a wider therapeutic window.[1]
It is important to acknowledge an ongoing pharmacological debate regarding the precise nature of this selectivity. Some evidence suggests that oliceridine's observed effects may be attributable to its behavior as a partial agonist with low intrinsic efficacy, rather than true G protein pathway-specific bias.[14] In this model, oliceridine's lower maximal stimulation of the receptor is sufficient to fully engage the highly amplified G protein pathway but is insufficient to trigger the threshold of receptor phosphorylation required for robust β-arrestin recruitment. While the "biased agonist" paradigm drove the drug's development, its clinical profile may be functionally indistinguishable from that of a partial agonist.[27] Regardless of the precise molecular mechanism, the intended and observed clinical outcome is a pharmacological profile that, at equianalgesic concentrations, may be associated with a reduced burden of certain adverse events compared to conventional full MOR agonists like morphine.
The pharmacokinetic profile of oliceridine is characterized by its intravenous route of administration, rapid distribution, hepatic metabolism into inactive compounds, and a relatively short half-life, befitting its role in managing acute pain. A summary of key parameters is provided in Table 2.
Table 2: Key Pharmacokinetic Parameters of Oliceridine
Parameter | Value | Clinical Implication |
---|---|---|
Tmax (IV) | < 5 minutes | Rapid onset of analgesia suitable for acute pain. |
Volume of Distribution (Vd) | 90–126 L | Moderate tissue distribution. |
Protein Binding | ~77% | Moderate binding to plasma proteins. |
Half-life (t1/2) - Parent | 1.3–3 hours | Short duration of action necessitates frequent dosing (e.g., PCA) for sustained analgesia. |
Half-life (t1/2) - Metabolites | ~44 hours | Metabolites are inactive, so their long half-life is not clinically significant. |
Clearance | 34–59.6 L/h | Rapid clearance from the body. |
Primary Metabolizing Enzymes | CYP3A4, CYP2D6 | Potential for drug-drug interactions with inhibitors or inducers of these enzymes. |
Route of Elimination | ~70% Renal, ~30% Fecal | Predominantly renal excretion of inactive metabolites. |
The regulatory pathway for oliceridine was notable for its initial challenges, which ultimately shaped the drug's final approved labeling and restrictions. Following the submission of a New Drug Application (NDA) by Trevena, Inc. in late 2017, the FDA's Anesthetic and Analgesic Drug Products Advisory Committee convened in October 2018.[19] The committee voted narrowly, 8 to 7, against recommending approval, citing concerns that the demonstrated benefits of oliceridine did not outweigh its risks, with specific attention given to its potential for QTc interval prolongation.[15] Subsequently, in November 2018, the FDA issued a Complete Response Letter, formally declining the application.[19]
After conducting additional safety analyses and engaging in further discussions with the agency, Trevena resubmitted the NDA in February 2020.[19] This revised application included a key risk mitigation strategy: a maximum recommended daily dose. On August 7, 2020, the FDA granted approval for oliceridine.[19] The approved indication is for the "management of acute pain in adults severe enough to require an intravenous opioid analgesic and for whom alternative treatments are inadequate".[1] This indication positions oliceridine as a second-line agent for a specific patient population in a specific clinical context.
The approval of oliceridine is accompanied by stringent limitations on its use, designed to ensure patient safety. It is indicated exclusively for short-term intravenous administration and must be used only in hospitals or other controlled clinical settings, such as during inpatient or outpatient procedures.[14] The drug is explicitly not approved for at-home or outpatient prescription use.[14]
A central element of its risk management is the maximum recommended cumulative daily dose of 27 mg.[14] This dose cap was established to mitigate the risk of QTc interval prolongation, the key safety concern that initially hindered its approval.[15] The imposition of this daily limit fundamentally defines oliceridine's clinical role as an agent for acute, short-duration pain management where total drug exposure is carefully controlled.
Reflecting its mechanism as a potent MOR agonist with demonstrated abuse potential comparable to other opioids, the U.S. Drug Enforcement Administration (DEA) has placed oliceridine in Schedule II of the Controlled Substances Act.[1] This classification imposes the strictest level of control for a medically approved substance, mandating specific prescribing, dispensing, and record-keeping practices to prevent diversion and misuse.
The efficacy and safety of oliceridine were primarily established in a robust Phase III program involving over 1,500 patients.[29] The program was anchored by two pivotal, randomized, double-blind, placebo- and active-controlled trials:
In both trials, patients were randomized to receive placebo, morphine, or one of three oliceridine dosing regimens administered via patient-controlled analgesia (PCA).[8] The primary efficacy endpoint was the responder rate, a categorical outcome defined as a patient achieving at least a 30% improvement from baseline in the time-weighted Sum of Pain Intensity Differences (SPID) without early discontinuation.[31] The program also included
ATHENA (NCT02656875), a large, open-label safety study that enrolled a broad population of 768 patients with moderate-to-severe acute pain from various surgical procedures or medical conditions, including elderly, obese, and medically complex patients, providing data on real-world use.[7]
The results from the APOLLO trials consistently demonstrated the analgesic efficacy of oliceridine.
The ATHENA open-label safety study provided supportive evidence of oliceridine's effectiveness in a more heterogeneous patient population than that included in the tightly controlled pivotal trials.[7] This study included patients with various comorbidities such as diabetes and sleep apnea, as well as elderly and obese individuals.[29] In this real-world setting, oliceridine, administered via clinician bolus, PCA, or a combination, was effective in providing rapid and sustained reduction in pain intensity scores from baseline.[7] The safety profile observed in ATHENA was consistent with the findings from the APOLLO trials, confirming its tolerability across a broad range of patients requiring intravenous opioid therapy.[7]
As a potent opioid agonist, oliceridine carries an FDA-mandated boxed warning, the most stringent warning for prescription drugs, highlighting several life-threatening risks common to the opioid class.[14]
Oliceridine is contraindicated in patients with:
The overall safety profile of oliceridine is similar to that of other intravenous opioids.[14] In the pivotal clinical trials, the most frequently reported treatment-emergent adverse events (occurring in ≥10% of patients) were nausea, vomiting, dizziness, headache, constipation, pruritus, and hypoxia.[33] The incidence and severity of these adverse events, particularly gastrointestinal and respiratory effects, were generally dose-dependent.[7] Most reported adverse events in the clinical program were of mild or moderate intensity.[7]
The clinical development program for oliceridine was designed with morphine as the active comparator, allowing for a direct comparison of efficacy and safety at equianalgesic doses. This comparative analysis is essential for defining oliceridine's potential role in clinical practice.
As established in the APOLLO trials, PCA demand doses of 0.35 mg and 0.5 mg of oliceridine were non-inferior to a 1 mg PCA demand dose of morphine for the management of postoperative pain.[7] This suggests an approximate relative potency ratio of 2-3:1 for PCA dosing (i.e., 1 mg of morphine is roughly equianalgesic to 0.35-0.5 mg of oliceridine). While non-inferiority was met, some analyses noted that morphine produced numerically higher response rates or greater pain reduction, though these differences were not statistically significant.[33]
The foundational hypothesis for oliceridine was that its G protein bias would translate into a significantly improved respiratory safety profile compared to conventional opioids. The preclinical data in animal models strongly supported this premise, suggesting a wider therapeutic window between analgesia and respiratory depression.[12] However, the results from the pivotal Phase III trials presented a more complex picture.
The APOLLO trials utilized a pre-specified secondary endpoint known as the Respiratory Safety Burden (RSB), a composite measure reflecting the cumulative duration of clinically relevant respiratory safety events (e.g., low respiratory rate, low oxygen saturation).[8] While oliceridine demonstrated a dose-dependent increase in RSB, the primary analysis
failed to show a statistically significant difference in RSB between the equianalgesic oliceridine regimens (0.35 mg and 0.5 mg) and the morphine 1 mg regimen.[7] The incidence of individual respiratory events was numerically lower in the oliceridine groups, but the trials may have been underpowered to detect a statistically significant difference for this endpoint.[35]
In contrast, subsequent exploratory, post-hoc analyses of the pooled trial data, as well as retrospective chart review studies, have suggested a more favorable respiratory profile. These analyses have reported a significantly lower incidence of operationally defined opioid-induced respiratory depression (OIRD) in patients treated with oliceridine compared to those treated with morphine or other conventional opioids.[12] While these findings are encouraging and align with the drug's proposed mechanism, they are not as robust as the results from the pre-specified analyses of the registration trials. Therefore, while there is a signal for improved respiratory safety, it was not definitively proven in the pivotal studies.
The most consistent and statistically robust advantage of oliceridine over morphine observed in the clinical program is its superior gastrointestinal tolerability profile. Postoperative nausea and vomiting (PONV) are highly prevalent and distressing adverse events that can delay recovery and increase healthcare costs.[25]
Across both APOLLO trials, patients treated with equianalgesic doses of oliceridine experienced significantly lower rates of nausea and vomiting and had a significantly lower requirement for rescue antiemetic medications compared to patients treated with morphine.[7] To quantify this benefit, an exploratory endpoint of "complete GI response"—defined as experiencing no vomiting and requiring no rescue antiemetics—was analyzed. After controlling for the level of analgesia, patients receiving oliceridine had 2 to 3 times higher odds of achieving a complete GI response compared to those receiving morphine (
p<0.05).[9] This clear and clinically meaningful benefit in GI tolerability represents the primary safety differentiator for oliceridine based on the highest level of clinical evidence.
Despite the novel mechanism of action, studies designed to evaluate abuse potential have concluded that oliceridine's profile is similar to that of other potent MOR agonists. A human abuse potential study directly comparing intravenous oliceridine to morphine found that equianalgesic doses produced similar subjective ratings on scales such as "Drug Liking," "Take Drug Again," and "Overall Drug Liking".[3] Preclinical studies in animal models of drug self-administration also demonstrated that oliceridine functions as a reinforcer with a potency and effectiveness comparable to that of morphine and oxycodone.[11] These findings strongly indicate that oliceridine retains a high potential for abuse and psychological dependence, fully justifying its placement as a Schedule II controlled substance. The potential for physical dependence is also assumed to be similar to other opioids.
Table 3: Comparative Summary of Oliceridine vs. Morphine in Pivotal Trials (APOLLO-1 & -2)
Endpoint | Oliceridine 0.35 mg PCA | Oliceridine 0.5 mg PCA | Morphine 1.0 mg PCA | Placebo | Key Finding/P-value |
---|---|---|---|---|---|
Responder Rate | Superior to Placebo | Superior to Placebo | Superior to Placebo | Baseline | Oliceridine regimens were statistically superior to placebo (p<0.05).8 |
Non-inferiority vs. Morphine | Non-inferior | Non-inferior | - | - | 0.35 mg and 0.5 mg oliceridine regimens were non-inferior to morphine.7 |
Respiratory Safety Burden (RSB) | Numerically Lower | Numerically Lower | Higher | Lower | No statistically significant difference between oliceridine and morphine regimens.7 |
Incidence of Nausea | Lower | Lower | Higher | Lower | Significantly lower relative risk with oliceridine regimens vs. morphine.8 |
Incidence of Vomiting | Lower | Lower | Higher | Lower | Significantly lower relative risk with oliceridine regimens vs. morphine.8 |
Use of Rescue Antiemetics | Lower | Lower | Higher | Lower | Odds ratio for rescue use was significantly lower in all oliceridine groups vs. morphine (p<0.05).11 |
"Complete GI Response" | Higher | Higher | Lower | Higher | Odds ratio for complete GI response was 2-3 times higher for oliceridine vs. morphine (p<0.05).9 |
The administration of oliceridine must be individualized based on the severity of pain, patient response, prior analgesic experience, and risk factors for adverse events. It is for intravenous use only and should be administered as part of a multimodal analgesic regimen whenever possible.[32]
Dosage adjustments and increased monitoring may be necessary for certain patient populations.
Oliceridine is a specialized agent, not a first-line replacement for all intravenous opioids. Its clinical profile suggests a specific niche in the management of moderate-to-severe acute pain in the controlled, inpatient setting. Its primary, evidence-backed advantage is a superior gastrointestinal tolerability profile compared to morphine, making it a compelling option for patients at high risk for PONV or for whom PONV could significantly complicate recovery or delay discharge. Its rapid onset of action is also advantageous in the immediate postoperative period.
The pharmacokinetic profile, particularly the lack of active metabolites, makes oliceridine a theoretically safer choice than morphine in patients with renal impairment, as the risk of toxic metabolite accumulation is eliminated. However, clinicians must remain vigilant. The anticipated revolutionary benefit in respiratory safety was not borne out in the pivotal trials, and its abuse potential is equivalent to that of morphine. The strict 27 mg daily dose limit, implemented to manage cardiac risk, firmly restricts its use to short-term scenarios (typically ≤48 hours). Therefore, oliceridine should be used with the same precautions and monitoring as any other potent Schedule II opioid, with its selection guided by a patient-specific assessment of the risks and benefits, particularly the trade-off between improved GI tolerability and the need for careful dose limitation.
Oliceridine (Olinvyk) marks an important, albeit incremental, advancement in opioid pharmacology. As the first clinically approved G protein-biased agonist of the μ-opioid receptor, it validates a novel drug design strategy aimed at separating analgesia from opioid-related adverse events. Clinical evidence from its robust development program confirms that it is an effective intravenous analgesic for moderate-to-severe acute pain, with an efficacy non-inferior to that of morphine.
The principal clinical advantage of oliceridine, supported by the highest level of evidence from its registration trials, is its significantly improved gastrointestinal safety profile, manifesting as a lower incidence of postoperative nausea and vomiting compared to morphine at equianalgesic doses. This positions oliceridine as a valuable therapeutic option for specific patient populations where minimizing PONV is a clinical priority. Furthermore, its metabolism into inactive compounds and lack of required dose adjustment in renal impairment offer a clear pharmacokinetic advantage over morphine.
However, the initial promise of a substantially safer opioid has been tempered by the clinical trial results. The hypothesized major improvement in respiratory safety was not statistically confirmed in the primary analyses of pivotal studies, and its abuse liability is comparable to that of conventional opioids. Its use is appropriately restricted to controlled clinical settings and is governed by a strict 27 mg maximum daily dose to mitigate the risk of QTc prolongation.
In conclusion, oliceridine is not a panacea for the opioid crisis nor a universally "safer" opioid. Rather, it is a specialized tool in the perioperative and acute pain armamentarium. Its rational use should be selective, targeted toward short-term (≤48 hours) scenarios in patients who may derive particular benefit from its favorable gastrointestinal and pharmacokinetic profiles. The introduction of oliceridine encourages a more nuanced, mechanism-informed approach to opioid selection, while simultaneously reinforcing the fundamental principles of cautious dosing, vigilant monitoring, and multimodal analgesia that are paramount to the safe and effective management of acute pain.
Published at: August 28, 2025
This report is continuously updated as new research emerges.
Empowering clinical research with data-driven insights and AI-powered tools.
© 2025 MedPath, Inc. All rights reserved.