2,578
Views
0
CrossRef citations to date
0
Altmetric
Position Statement

American College of Medical Toxicology and the American Academy of Clinical Toxicology position statement: nalmefene should not replace naloxone as the primary opioid antidote at this time

ORCID Icon, ORCID Icon, ORCID Icon & ORCID Icon
Pages 952-955 | Received 12 Oct 2023, Accepted 09 Nov 2023, Published online: 01 Dec 2023

Abstract

Background

Nalmefene is a potent opioid antagonist that has recently been reintroduced in the United States to treat known or suspected opioid overdose.

Nalmefene clinical trial data

The injection formulation, which had been withdrawn in 2008, was reintroduced in 2022, and in 2023 the United States Food and Drug Administration approved a new intranasal formulation of nalmefene. Because nalmefene had been previously approved for use in 1995 via injection, the new intranasal formulation did not require new clinical data as it was approved under an Abbreviated New Drug Application. Inherent to this abbreviated approval process, intranasal nalmefene was not studied in patients currently suffering opioid overdose.

Naloxone and nalmefene

Nalmefene also has unique characteristics compared with naloxone, the current standard opioid antidote. Nalmefene has a higher affinity for opioid receptors and a longer duration of action than naloxone. Comparative effectiveness data regarding naloxone and nalmefene are sparse, and it is unclear if the inherent properties of nalmefene are beneficial in opioid overdose. We have decades of experience using naloxone safely and effectively as the primary opioid antidote, even in cases of fentanyl and fentanyl analog overdoses. There is, however, evidence to suggest nalmefene may result in more prolonged and severe opioid withdrawal than naloxone, which could be harmful to patients.

Position

As nalmefene is untested in the current clinical environment of synthetic opioid overdoses and has the potential to cause harm via prolonged withdrawal, it is the opinion of the American College of Medical Toxicology and the American Academy of Clinical Toxicology that nalmefene should not replace naloxone as the primary opioid antidote at this time.

Recommendations

We recommend additional clinical studies of nalmefene, administered via all approved routes, be conducted in a comparative fashion with naloxone, and that safety and effectiveness outcomes be evaluated before nalmefene is recommended as a primary opioid antidote.

View correction statement:
Correction

Background

Against the backdrop of a severe opioid crisis that resulted in over 80,000 deaths in the United States in 2021, with synthetic opioids being a primary culprit, researchers are exploring alternative treatment approaches [Citation1]. On May 22, 2023, the United States Food and Drug Administration (US FDA) approved nalmefene nasal spray (OPVEE®, Indivior®) for emergency treatment of opioid overdose [Citation2].

Although this is a newly approved route of administration, nalmefene is not a new drug. Nalmefene is an opioid antagonist that was originally US FDA-approved for injection (via the intravenous, intramuscular, or subcutaneous routes) in 1995 [Citation3]. The original injectable nalmefene product was removed from the market for commercial reasons in 2008 [Citation4,Citation5]. Clinicians had not found a consistent role for nalmefene, presumably because it did not have a clinically relevant duration of action or ease of use. Naloxone, originally US FDA-approved in 1971 [Citation6], continues to be recognized as the leading opioid antagonist by all routes of administration. Because nalmefene has a high affinity for opioid receptors and a long duration of action compared to naloxone, it is marketed as advantageous for use following an overdose of synthetic opioids, despite the lack of supporting clinical effectiveness and safety data [Citation7–9].

Because nalmefene was already approved by the US FDA for use via intramuscular injection, the intranasal product did not require new clinical data [Citation9–11]. The intranasal nalmefene product was submitted to the US FDA as part of an Abbreviated New Drug Application pathway, which required that the sponsor only demonstrate the new administration route is bioequivalent and pharmacokinetically equivalent to the older injectable product [Citation9,Citation11,Citation12].

Nalmefene clinical trial data

Nalmefene originally received US FDA approval in 1995 (Revex®) via injection based on data from controlled trials on a total of 1,127 patients involving either postoperative opioid reversal or opioid overdose [Citation3]. The original prescribing information and US FDA approval letter provide data on opioid poisoning limited to 284 patients from four trials involving patients presumed to have taken an opioid overdose [Citation3]. While the Revex® product was withdrawn in 2008, US FDA determined it was not withdrawn for reasons of safety or effectiveness, allowing for future approvals through an Abbreviated New Drug Application [Citation13]. The prescribing information for the newly US FDA-approved generic parenteral formulation of nalmefene contains data from the same 1,127 patients (again, 284 of whom were presumed to have experienced an opioid overdose), while the prescribing information of the new intranasal formulation reviews these same 1,127 patients solely for adverse events [Citation8,Citation9]. The prescribing information of neither formulation provides new data gathered from patients with opioid overdose reversed with nalmefene.

Pharmacokinetic data used in the Abbreviated New Drug Application to obtain US FDA-approval via the intranasal route were obtained after intranasal nalmefene was administered to volunteers under controlled conditions and not in patients currently suffering from an opioid overdose.

Clinical data on nalmefene used to safely reverse opioid poisoning are sparse [Citation14]. Only one double-blind controlled trial comparing naloxone to nalmefene has been published, and it examined the intravenous route for both drugs [Citation15]. This trial, which randomized 176 patients to one of three treatments (naloxone 2 mg, nalmefene 1 mg, or nalmefene 2 mg), found all three treatments to be effective and safe. While this study found no difference in adverse effects between treatment arms, this study was likely underpowered to detect differences in adverse effects, which were approximately twice as common (31%) with nalmefene 2 mg compared to nearly 16% for both nalmefene 1 mg and naloxone 2 mg. While data from this study are used in support of intranasal nalmefene in the Abbreviated New Drug Application, it is notable that the authors of this study warn in their manuscript “Clinicians concerned about possible prolonged withdrawal or adverse reactions to nalmefene may want to try naloxone first.” Importantly, this trial was conducted in the 1990s, long before fentanyl became the most common cause of opioid overdose in the US. While parenteral nalmefene, like naloxone, reverses the effects of fentanyl in volunteer studies [Citation16,Citation17], unlike naloxone, data on nalmefene reversal of fentanyl overdose via the intranasal route are lacking.

Naloxone and nalmefene

Naloxone is shorter-acting and is US FDA-approved for administration by intramuscular, intravenous, intranasal and subcutaneous routes. Compared to naloxone, nalmefene has a 5-fold higher binding affinity for opioid receptors (measured by receptor affinity, Ki) than naloxone [Citation6,Citation18]. (See ).

Table 1. Comparison of naloxone and nalmefene.

The duration of action of nalmefene compared to naloxone has been promoted as beneficial [Citation7], presumably because it may reduce the use of naloxone infusions or repeat naloxone administration. The primary adverse effect of any opioid antagonist, naloxone included, is to cause acute withdrawal in patients with opioid dependence. Precipitated opioid withdrawal can range in severity from mild dysphoria or gastrointestinal upset to life-threatening conditions including acute respiratory distress syndrome, severe agitation, dysrhythmias, and stress cardiomyopathy. The longer duration of action of nalmefene may predispose to a lengthier period of precipitated opioid withdrawal.

The longer duration of action of nalmefene does not eliminate the need for medical observation after administration. We are concerned that the long duration of action may provide a false sense of comfort that no further care is needed. Compared to naloxone, a long-acting antagonist may result in increased patient emergency department length of stay and resource utilization. Individuals reversed with nalmefene in the out-of-hospital environment who do not seek formal medical care may attempt to self-treat withdrawal with opioids. The current standard is to observe patients for recurrent sedation and respiratory depression after the opioid antagonist effect dissipates- approximately 90 min following intravenous naloxone [Citation19]. Patients who receive nalmefene may require longer periods of observation, up to several hours, to observe for resedation and recurrent respiratory depression as the longer-acting antagonist effects wane.

It is not clear that an antagonist with a higher affinity for the opioid receptor is desirable or would result in better outcomes, even in patients with synthetic opioid overdose. Increased affinity may contribute to more severe and intractable withdrawal. We do not have clinical data for nalmefene in patients with synthetic opioid overdose.

Moreover, the current standard opioid antidote, naloxone, has a sufficiently high opioid receptor affinity to reverse novel synthetic opioids. In a study of volunteers given the high-affinity synthetic opioid carfentanil, administration of naloxone 2 mg intravenously blocked 80.6% of receptors at 5 min [Citation20]. Although nalmefene 1 mg intravenously blocked 99% of receptors, the 80% occupancy produced by naloxone is sufficient to restore ventilation, which is the clinical goal. In current clinical practice, much lower doses of naloxone are initially administered to blunt the risk and severity of precipitated opioid withdrawal, highlighting the limited benefit of a higher affinity reversal agent. In published case reports, naloxone successfully reversed dozens of cases of high-dose fentanyl and fentanyl analog overdoses, though on occasion higher or titrated doses were used [Citation21–23]. A single-center retrospective review found no difference in naloxone dose in patients with fentanyl compared to heroin overdose [Citation24]. We are unable to find clinical evidence that a strategy of repeated or escalating naloxone doses is inferior to a longer-acting antagonist for the treatment of opioid overdose.

Position

As physicians, pharmacists, scientists, and specialists in poison information, we are experts in pharmacology, toxicology, and the management of opioid overdose and addiction. We applaud the effort to seek out new therapeutic strategies for the management of these patients.

We are concerned that the use of a longer-acting reversal agent would not improve on current practice and could potentially cause harm. When withdrawal is precipitated by an opioid antagonist, there are few good management options. In most cases, the best strategy is to address and support the patient’s signs and symptoms until the effects of the antagonist wane. In the case of naloxone, which has a relatively short duration of action, severe withdrawal usually lasts less than an hour with symptoms typically persisting no more than 90 min [Citation25–27]. A longer-acting antagonist is anticipated to cause longer-lasting precipitated withdrawal and may lead to worse patient outcomes. Clinical experience with both naltrexone and nalmefene suggests prolonged withdrawal is a complication of longer-acting opioid antagonists [Citation28]. Although a longer-acting antagonist may be theoretically beneficial for the resuscitation of opioid-naive individuals in an opioid-induced mass casualty incident, this type of event has never been reported in North America and this application is unstudied.

We are also concerned that patients who receive nalmefene may require longer periods of observation, by up to several hours, to observe for recrudescent effects as the antagonist effects wane. Patients who receive nalmefene will still need medical observation to ensure that respiratory depression does not recur after the effects of the medication subside. This will prolong emergency department visit length and challenge patient and clinician expectations, further burdening a taxed system. Further clinical study is needed to understand whether a reduction in repeat antagonist use justifies a longer length of stay or longer period of withdrawal.

Finally, we are concerned that intranasal nalmefene has not been adequately studied for effectiveness in the actual setting and patient population: for patients with severe opioid intoxication in the out-of-hospital environment. Lack of proof of safety and efficacy in real-world use could result in significant harm if widely utilized.

The potential benefits of nalmefene over naloxone (greater opioid receptor affinity, longer duration action) carry the risk of causing harm. These benefits, if present, should be demonstrated in the clinical environment, balanced with the risks, and compared to naloxone prior to the broad adoption of nalmefene.

Recommendations

We recommend the following actions to inform safe and effective opioid reversal:

  1. Continue to recommend naloxone as the preferred first-line agent until, and if, more robust clinical and cost data are available to support the routine use of nalmefene.

  2. Conduct additional clinical studies of nalmefene (via the intravenous, intramuscular and intranasal routes) to determine the effectiveness of the drug in its anticipated clinical setting (overdose patients in hospital and out-of-hospital environments).

  3. Evaluate important safety endpoints for nalmefene use, particularly related to complications of opioid reversal, such as acute respiratory distress syndrome and prolonged precipitated withdrawal.

  4. Perform comparative studies with naloxone to determine differences in effectiveness, adverse outcomes, effect on emergency department length of stay and other relevant clinical measures, effect on initiation of medications for opioid use disorder, medication and healthcare cost and overall resource utilization.

Disclaimers

While individual practices may differ, this is the position of the American College of Medical Toxicology and American Academy of Clinical Toxicology at the time written, after a review of the issue and pertinent literature.

The American College of Medical Toxicology Receives funding from the US FDA to study opioid antagonists. This statement does not necessarily represent the official views, nor an endorsement, by the US FDA/US Department of Health and Human Services, or the US Government.

Disclosure statement

No potential conflict of interest was reported by the authors.

Correction Statement

This article was originally published with errors, which have now been corrected in the online version. Please see Correction (http://dx.doi.org/10.1080/15563650.2023.2293385)

Additional information

Funding

The authors reported there is no funding associated with the work featured in this article.

References

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.