Comment on: Can Intranasal Nalmefene Reduce the Number of Opioid Overdose Deaths?

IF 1.5 4区 医学 Q3 PHARMACOLOGY & PHARMACY Clinical Pharmacology in Drug Development Pub Date : 2024-01-30 DOI:10.1002/cpdd.1382
Phil Skolnick
{"title":"Comment on: Can Intranasal Nalmefene Reduce the Number of Opioid Overdose Deaths?","authors":"Phil Skolnick","doi":"10.1002/cpdd.1382","DOIUrl":null,"url":null,"abstract":"<p>With an emphasis on the disappointing limited impact of naloxone in reducing opioid overdose deaths, Sellers and Romach<span><sup>1</sup></span> raise a number of compelling arguments. Their view is supported by data from the Centers for Disease Control and Prevention: According to the State Unintentional Drug Overdose Reporting System database, naloxone had been administered in more than 20% of opioid-related overdose deaths nationally, with some jurisdictions reporting that more than 35% of decedents had received naloxone.<span><sup>2</sup></span> Thus, the estimated death toll from opioid overdoses exceeded 85,000 for the 12-month period ending July 2023<span><sup>3</sup></span> despite the availability of multiple naloxone products and naloxone saturation strategies.<span><sup>4</sup></span> More than 90% of these fatalities were attributed to high-potency synthetic opioids (synthetics) such as fentanyl.<span><sup>3</sup></span></p><p>In addition to the points raised by Sellers and Romach,<span><sup>1</sup></span> these data raise another fundamental issue: Is the availability of standard naloxone tools (ie, doses of 4 mg intranasal and 2 mg/2 mL intramuscular) available in a community setting sufficient to effectively reverse a synthetic opioid overdose?</p><p>There is now compelling evidence<span><sup>5-8</sup></span> that rapid delivery of higher doses of naloxone are needed to reverse an overdose involving synthetics compared to morphinan-based molecules such as morphine and heroin. Synthetics have a more rapid onset of action than morphinans,<span><sup>5, 6</sup></span> which reduces the window of opportunity to successfully reverse an overdose. Both the shorter time to maximum concentration (t<sub>max</sub>; 0.25 hours) and higher maximum concentration (C<sub>max</sub>; 12.2 ng/mL)<span><sup>9</sup></span> of intranasal nalmefene (2.7 mg), when taken together with a higher affinity at μ-opioid receptors,<span><sup>6</sup></span> are consistent with a more rapid onset of action compared to intranasal naloxone (t<sub>max</sub> 0.5 hours and C<sub>max</sub> 5.3 ng/mL, respectively).<span><sup>6</sup></span> However, in an overdose involving synthetics, antagonist exposure during the crucial first few minutes after dosing is far more significant<span><sup>7</sup></span>: In this regard, plasma nalmefene concentrations 5 minutes after administration<span><sup>9</sup></span> are approximately 3-fold higher than following intranasal naloxone.<span><sup>6</sup></span> A more rapid onset of action is also supported by comparative efficacy data in a clinical model of opioid-induced respiratory depression.<span><sup>10</sup></span> In this study, intranasal nalmefene led to a reversal of remifentanil-induced reductions in minute ventilation that was almost twice as high as the reversal produced by intranasal naloxone (5.75 vs 3.01 L/min; <i>P</i> less than .0009) at 5 minutes after administration, the primary end point).</p><p>The dose of naloxone required to reverse an overdose is symptom driven and, by this criterion, empirical. Multiple factors, ranging from the type and quantity of opioid and the presence of other drugs (eg, alcohol, benzodiazepines) to the victim's general health, challenge accurate analysis of the quantities of naloxone required to reverse an overdose.<span><sup>6</sup></span> Nonetheless, multiple clinical reports indicate higher doses of naloxone are required to reverse an overdose involving synthetics, with some authors recommending parenteral administration of 12-15 mg.<span><sup>6</sup></span> Furthermore, in a well-controlled preclinical study that eliminates the many variables attending an overdose, Kelly et al<span><sup>5</sup></span> have demonstrated that a 10-fold higher dose of naloxone is required to reverse fentanyl- compared to morphine-induced respiratory depression. In a quantitative translational approach, Mann et al<span><sup>7</sup></span> recently developed and validated a model to evaluate the effectiveness of intramuscular naloxone in reversing the hypoxia-induced cardiac arrest produced by fentanyl and carfentanil. In the absence of intervention, an intravenous bolus of 1.63 mg fentanyl resulted in cardiac arrest in approximately 52% of 2000 virtual patients in this model. An intramuscular dose of naloxone (2 mg/2 mL) favored by many first responders reduced the incidence of cardiac arrest to 30% in model simulations of 2000 patients, a rescue rate of approximately 42%. We have implemented and expanded this model to compare the effectiveness of intranasal nalmefene to intranasal naloxone (4 mg), generally viewed as the “gold standard” in a community setting. Nalmefene resulted in clinically meaningful reductions in the incidence of cardiac arrest compared to intranasal naloxone at fentanyl and carfentanil doses that resulted in a 50%-90% incidence of cardiac arrest in the absence of antagonist treatment (manuscript in preparation).</p><p>Many of the important issues raised by Sellers and Romach<span><sup>1</sup></span> reflect a broader societal failure to effectively destigmatize and treat opioid use disorder. Effective deployment of reversal agents is only one facet of a strategy to mitigate the societal impact of opioid misuse. Nonetheless, based on the available data, adoption of nalmefene nasal spray in a community use setting, where establishing an intravenous line is not practical and respiratory support is often absent, will have a significant impact on reducing overdose deaths in the synthetic opioid era.</p><p>The author is an employee of Indivior Inc. and is named as a coinventor on four patent applications which describe formulations, compositions, and methods for the prevention and treatment of opioid overdose.</p>","PeriodicalId":10495,"journal":{"name":"Clinical Pharmacology in Drug Development","volume":null,"pages":null},"PeriodicalIF":1.5000,"publicationDate":"2024-01-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cpdd.1382","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical Pharmacology in Drug Development","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cpdd.1382","RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q3","JCRName":"PHARMACOLOGY & PHARMACY","Score":null,"Total":0}
引用次数: 0

Abstract

With an emphasis on the disappointing limited impact of naloxone in reducing opioid overdose deaths, Sellers and Romach1 raise a number of compelling arguments. Their view is supported by data from the Centers for Disease Control and Prevention: According to the State Unintentional Drug Overdose Reporting System database, naloxone had been administered in more than 20% of opioid-related overdose deaths nationally, with some jurisdictions reporting that more than 35% of decedents had received naloxone.2 Thus, the estimated death toll from opioid overdoses exceeded 85,000 for the 12-month period ending July 20233 despite the availability of multiple naloxone products and naloxone saturation strategies.4 More than 90% of these fatalities were attributed to high-potency synthetic opioids (synthetics) such as fentanyl.3

In addition to the points raised by Sellers and Romach,1 these data raise another fundamental issue: Is the availability of standard naloxone tools (ie, doses of 4 mg intranasal and 2 mg/2 mL intramuscular) available in a community setting sufficient to effectively reverse a synthetic opioid overdose?

There is now compelling evidence5-8 that rapid delivery of higher doses of naloxone are needed to reverse an overdose involving synthetics compared to morphinan-based molecules such as morphine and heroin. Synthetics have a more rapid onset of action than morphinans,5, 6 which reduces the window of opportunity to successfully reverse an overdose. Both the shorter time to maximum concentration (tmax; 0.25 hours) and higher maximum concentration (Cmax; 12.2 ng/mL)9 of intranasal nalmefene (2.7 mg), when taken together with a higher affinity at μ-opioid receptors,6 are consistent with a more rapid onset of action compared to intranasal naloxone (tmax 0.5 hours and Cmax 5.3 ng/mL, respectively).6 However, in an overdose involving synthetics, antagonist exposure during the crucial first few minutes after dosing is far more significant7: In this regard, plasma nalmefene concentrations 5 minutes after administration9 are approximately 3-fold higher than following intranasal naloxone.6 A more rapid onset of action is also supported by comparative efficacy data in a clinical model of opioid-induced respiratory depression.10 In this study, intranasal nalmefene led to a reversal of remifentanil-induced reductions in minute ventilation that was almost twice as high as the reversal produced by intranasal naloxone (5.75 vs 3.01 L/min; P less than .0009) at 5 minutes after administration, the primary end point).

The dose of naloxone required to reverse an overdose is symptom driven and, by this criterion, empirical. Multiple factors, ranging from the type and quantity of opioid and the presence of other drugs (eg, alcohol, benzodiazepines) to the victim's general health, challenge accurate analysis of the quantities of naloxone required to reverse an overdose.6 Nonetheless, multiple clinical reports indicate higher doses of naloxone are required to reverse an overdose involving synthetics, with some authors recommending parenteral administration of 12-15 mg.6 Furthermore, in a well-controlled preclinical study that eliminates the many variables attending an overdose, Kelly et al5 have demonstrated that a 10-fold higher dose of naloxone is required to reverse fentanyl- compared to morphine-induced respiratory depression. In a quantitative translational approach, Mann et al7 recently developed and validated a model to evaluate the effectiveness of intramuscular naloxone in reversing the hypoxia-induced cardiac arrest produced by fentanyl and carfentanil. In the absence of intervention, an intravenous bolus of 1.63 mg fentanyl resulted in cardiac arrest in approximately 52% of 2000 virtual patients in this model. An intramuscular dose of naloxone (2 mg/2 mL) favored by many first responders reduced the incidence of cardiac arrest to 30% in model simulations of 2000 patients, a rescue rate of approximately 42%. We have implemented and expanded this model to compare the effectiveness of intranasal nalmefene to intranasal naloxone (4 mg), generally viewed as the “gold standard” in a community setting. Nalmefene resulted in clinically meaningful reductions in the incidence of cardiac arrest compared to intranasal naloxone at fentanyl and carfentanil doses that resulted in a 50%-90% incidence of cardiac arrest in the absence of antagonist treatment (manuscript in preparation).

Many of the important issues raised by Sellers and Romach1 reflect a broader societal failure to effectively destigmatize and treat opioid use disorder. Effective deployment of reversal agents is only one facet of a strategy to mitigate the societal impact of opioid misuse. Nonetheless, based on the available data, adoption of nalmefene nasal spray in a community use setting, where establishing an intravenous line is not practical and respiratory support is often absent, will have a significant impact on reducing overdose deaths in the synthetic opioid era.

The author is an employee of Indivior Inc. and is named as a coinventor on four patent applications which describe formulations, compositions, and methods for the prevention and treatment of opioid overdose.

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
评论鼻内注射纳美芬能减少阿片类药物过量致死的人数吗?
Sellers 和 Romach1 强调纳洛酮在减少阿片类药物过量死亡方面的作用有限,令人失望。他们的观点得到了美国疾病控制和预防中心数据的支持:根据 "州意外用药过量报告系统 "数据库的数据,全国 20% 以上与阿片类药物相关的用药过量死亡病例使用了纳洛酮,一些辖区报告称 35% 以上的死者使用了纳洛酮。因此,尽管有多种纳洛酮产品和纳洛酮饱和策略,截至 2023 年 7 月的 12 个月期间,阿片类药物过量造成的死亡人数估计仍超过 85,000 人3:现在有令人信服的证据5-8 表明,与吗啡和海洛因等吗啡类分子相比,合成类阿片过量时需要快速给药更大剂量的纳洛酮才能逆转。合成类药物的起效时间比吗啡类药物更快,5、6 这就减少了成功逆转用药过量的机会窗口。鼻内注射纳美芬(2.7 毫克)达到最大浓度的时间(tmax;0.25 小时)较短,最大浓度(Cmax;12.2 纳克/毫升)9 较高,同时对μ-阿片受体的亲和力6 较高,这表明与鼻内注射纳洛酮相比,纳美芬起效更快(tmax 0.5 小时,Cmax 5.6 然而,在合成药物用药过量的情况下,用药后关键的最初几分钟内的拮抗剂暴露量要大得多7:在这方面,用药后 5 分钟9 的血浆纳美芬浓度比鼻内注射纳洛酮后的浓度高出约 3 倍。6 在阿片类药物引起的呼吸抑制临床模型中的疗效比较数据也支持纳美芬起效更快。在该研究中,鼻内注射纳美芬可逆转瑞芬太尼诱导的分钟通气量减少,在给药后 5 分钟(主要终点),其逆转效果几乎是鼻内注射纳洛酮逆转效果的两倍(5.75 L/min vs 3.01 L/min;P 小于 0.0009)。从阿片类药物的类型和数量、是否存在其他药物(如酒精、苯二氮卓)到受害者的总体健康状况等多种因素,都对准确分析逆转用药过量所需的纳洛酮剂量提出了挑战。6 不过,多份临床报告显示,逆转合成药物用药过量需要更高的纳洛酮剂量,一些作者建议肠外给药 12-15 毫克。此外,在一项控制良好的临床前研究中,Kelly 等人5 排除了用药过量时的许多变数,结果表明,与吗啡诱导的呼吸抑制相比,逆转芬太尼所需的纳洛酮剂量要高出 10 倍。Mann 等人7 最近开发并验证了一个定量转化方法模型,用于评估肌肉注射纳洛酮在逆转芬太尼和卡芬太尼引起的缺氧性心脏骤停方面的效果。在没有干预的情况下,静脉注射 1.63 毫克芬太尼会导致该模型中 2000 名虚拟患者中约 52% 的人心跳骤停。在对 2000 名患者进行的模型模拟中,许多急救人员所青睐的纳洛酮肌肉注射剂量(2 毫克/2 毫升)将心脏骤停的发生率降至 30%,抢救率约为 42%。我们实施并扩展了这一模型,以比较鼻内注射纳美芬与鼻内注射纳洛酮(4 毫克)的效果,后者在社区环境中通常被视为 "黄金标准"。与芬太尼和卡芬太尼剂量下的鼻内纳洛酮相比,纳美芬能在临床上显著降低心脏骤停的发生率,而在没有拮抗剂治疗的情况下,芬太尼和卡芬太尼剂量导致心脏骤停的发生率为 50%-90%(手稿正在撰写中)。有效使用逆转剂只是减轻阿片类药物滥用的社会影响战略的一个方面。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
CiteScore
3.70
自引率
10.00%
发文量
154
期刊介绍: Clinical Pharmacology in Drug Development is an international, peer-reviewed, online publication focused on publishing high-quality clinical pharmacology studies in drug development which are primarily (but not exclusively) performed in early development phases in healthy subjects.
期刊最新文献
Population Pharmacokinetics Modeling and Simulation of Deutenzalutamide, A Novel Androgen Receptor Antagonist, in Patients With Metastatic Castration-Resistant Prostate Cancer. Bioequivalence Study of Velpatasvir/Sofosbuvir Oral Coated Tablets in Healthy Volunteers Under Fasting Conditions. A Randomized, Cross-Over Study Investigating the Comparability of Somatrogon-ghla in 2 Different Drug Product Presentations. Bioequivalence of Recombinant Human Teriparatide Injection in Healthy Adult Female Subjects in the Fasting State. Effect of Erdafitinib on the Pharmacokinetics of Midazolam and Metformin in Patients With Advanced Solid Tumors Harboring FGFR Gene Alterations.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1