Expert Opinion on Drug Metabolism & Toxicology ISSN: 1742-5255 (Print) 1744-7607 (Online) Journal homepage: https://www.tandfonline.com/loi/iemt20 Safety and performance of current abusedeterrent formulations Rand Ahmad, Samaneh Alaei & Hamid Omidian To cite this article: Rand Ahmad, Samaneh Alaei & Hamid Omidian (2018) Safety and performance of current abuse-deterrent formulations, Expert Opinion on Drug Metabolism & Toxicology, 14:12, 1255-1271, DOI: 10.1080/17425255.2018.1546289 To link to this article: https://doi.org/10.1080/17425255.2018.1546289 Published online: 02 Dec 2018. Submit your article to this journal Article views: 190 View related articles View Crossmark data Citing articles: 2 View citing articles Full Terms & Conditions of access and use can be found at https://www.tandfonline.com/action/journalInformation?journalCode=iemt20 EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 2018, VOL. 14, NO. 12, 1255–1271 https://doi.org/10.1080/17425255.2018.1546289 REVIEW Safety and performance of current abuse-deterrent formulations Rand Ahmad, Samaneh Alaei and Hamid Omidian College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA ARTICLE HISTORY ABSTRACT Introduction: Prescription opioid abuse is now an epidemic that has forced the government and industries to take initiatives. These include developing abuse-deterrent formulations (ADFs), issuing regulatory guidances and allocating massive budgets to ensure the safety and effectiveness of these medications. Areas covered: This review covers the regulatory guidance on evaluation and labeling of the branded and generic ADFs. It also includes the relevant patents and technologies, the in-vitro, in-vivo, the postmarketing data, the FDA reviews, and the products’ labeling of the FDA-approved products with abusedeterrent features. Expert opinion: Despite the development of a dozen products with abuse-deterrent features, most of these technologies rely on the same deterrent agent, making it easier for abusers to focus their manipulation efforts and share their experience to defeat the technology. Further advancement in the field requires developing more robust, more diverse, safer, and affordable deterrent technologies for the extended- and immediate-release opioid products. Moreover, advances in the reporting of the post-market results, issuance of policies in support of the ADFs, and concurrent monitoring of the illicit opioid market are other considerations that can further help in confronting the epidemic. 1. Introduction Opioid painkillers are analgesics used to manage severe pain such as lower back pain and pain experienced by cancer patients or patients undergoing major surgeries [1]. Misuse, abuse, overdose, subsequent health complications, and death have been associated with the use of this class of prescription analgesics, representing the prescription opioid epidemic. The emergence rate of new nonmedical use of prescription drugs estimated to be around 5,700 cases/day in 2013 [2]. In 2012 alone, 259 million prescriptions were filled [3], reflecting the ease of obtaining prescription drugs as a result of overprescribing [4]. Despite the later awareness about the addiction potential associated with opioid analgesics, more than half of the Americans in 2017 were prescribed opioids with even an increased number of days per prescription, 18 days on average [5]. The emergency rooms have also recorded a high number of visits related to the prescription opioid abuse, around 488,000 visits in 2011 [2]. According to the CDC, the sales of prescription opioid drugs have quadrupled from 1999 to 2014, and this was paralleled by increased overdose deaths. The deaths among young adults (18–25 years old) were reported to be more than 1,700 in 2014, mainly due to prescription opioid overdosing [6]. In the US alone, the number of deaths has increased over the years, exceeding 16,500 cases in 2016, again as a result of overdosing prescription opioids [7]. This number is reported to be five times higher than in 1999 [5]. In connection with the high number of deaths in 2016, the cases of prescription opioids misuse were reported to reach CONTACT Hamid Omidian omidian@nova.edu S University Drive, Fort Lauderdale, FL 33328, USA © 2018 Informa UK Limited, trading as Taylor & Francis Group Received 14 March 2018 Accepted 6 November 2018 KEYWORDS Opioids; abuse-deterrent; formulation; technology; post-marketing; safety; toxicity 11.5 million cases, out of which 2.1 million were the first time misuse [8]. The problem of abusing prescription opioids also affected the economy. In 2016, the White House proposed $1.1 billion to treat opioid-use disorder as part of tackling the epidemic of abusing illicit and prescription opioids [9]. In 2017, an overall cost of the prescription opioid abuse was estimated around $78.5 billion [10]. Prescription drug abuse continues to be a burden on the health care and the economy at a significant national scale, driving Federal government, pharmaceutical industries, academic institutions, and most recently insurance companies to combat this crisis. In response to the prescription opioid epidemic, a dozen of abuse-deterrent formulations (ADFs) have been developed. These formulations are equipped with technologies to resist the drug products’ manipulation and abuse by alternate routes (Figure 1) while maintaining the proper drug release when used as intended. Methods of manipulating the opioid products include crushing/chewing the tablets for oral administration, coingestion of the drug product with alcohol, oral administration of multiple units of the drug product, crushing the drug product into fine particles for intranasal (IN) administration, and crushing and dissolving the drug product in a solvent suitable for intravenous (IV) administration [11]. These methods of manipulation aim to achieve a faster drug release and rapid onset of euphoric effect. Among all, the IV drug administration may be considered the most dangerous route of abuse [12–14]. The drug is injected directly into the bloodstream, increasing the risk of overdose and death, as well as hepatitis C and HIV infections. Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, 3200 1256 R. AHMAD ET AL. Article highlights ● ● ● ● ● ● ● The number of deaths due to prescription opioid abuse was over 16,500 in 2016 with an estimated cost of ~ $78.5 billion in 2017. Prescription opioid abuse is a complicated problem, exposing abusers to subsequent overdose health complications as well as toxicities derived from the excipients, the impurities, and the degradants of the manipulated drug and excipients; in particular when taken at larger amounts than allowed and administered via a non-oral route. The currently marketed ADFs, in general, have shown promise in reducing prescription drug abuse based on various pre-market studies and the available post-market surveillance. Despite the promising outcomes, the two major challenges left to be tackled are the liability to abuse by multiple oral administration of intact dosage units and the limited availability of the post-market data. Safer, more robust, more diverse, and cheaper deterring technologies need to be developed for both the extended- and immediate-release dosage forms. The unlimited number of ways that a prescription opioid can be manipulated calls for the FDA to continuously revise its guidelines to better evaluate the abuse-deterrent capability of the submitted ADFs. Further success and promise with the ADFs can be reinforced by the Government and State policies in support of their development, prescription, and insurance coverage. This box summarizes key points contained in the article. In addition to developing ADFs to combat the opioid drug epidemic, the FDA has outlined guidelines for their in-vitro and in-vivo evaluations. Products are evaluated based on the anticipated routes of abuse along with the characteristics of the deterrent technology utilized in each product. There are currently 17 approved products (Table 1), some acquired the FDA abuse-deterrent labeling, while others failed to fully comply with the FDA requirements despite exhibiting some abusedeterrent features. This paper collectively reviews the FDA regulatory considerations in evaluating the ADFs and the status of the approved products, their formulation, and manufacturing technologies, Figure 1. Abuse-deterrent technologies used in the FDA-approved products. in-vitro and in-vivo testing, safety, and post-marketing data, where available. It is paramount to periodically review the available data for these products. This allows for the early detection of any flaws in their technologies and formulations as well as the associated challenges in the field, which may emerge over time (e.g., study design challenges, potential toxicities). 2. FDA considerations for evaluating and labeling ADFs Two guidances have been issued by the FDA [18,19], one for the branded products and another for the generics, both presenting general recommendations to assess the ADF products. 2.1. The FDA guidance for the branded ADFs The FDA guidance assessing the branded products was finalized in April 2015. It describes the various categories of the ADFs based on the tamper-resistant technologies that they utilize. In principle, these categories are either dependent on the formulation or on the new discoveries as described in Figure 1. In addition to the ADFs’ categories, the guidance describes the premarket and post-market studies required for their evaluations. The pre-market studies involve three categories of testing; the invitro manipulation studies, the in-vivo pharmacokinetic studies, and the in-vivo clinical abuse potential studies. The post-market studies include a formal epidemiological surveillance as well as supportive studies. Table 2 provides a full description of the preand post-market studies. The study design should take into consideration the formulation, the abuse-deterrent technology, all the anticipated routes of abuse, the appropriate positive controls, comparators, placebos, and the results of the earlier studies (e.g., the best extracting solvent determined from the category 1 studies should be used in the category 2 pharmacokinetic studies). EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 1257 Table 1. Summary of the products with abuse-deterrent features. Product name Arymo ER Embeda Exalgo Hysingla ER Nucynta ER Morphabond ER Opana ER Oxaydo Deterring technology Guardian™: A crush and extraction resistant, injection- molded matrix Agonist/Antagonist: A sequestered antagonist OROS: A crush resistant semipermeable membrane and an extraction resistant core RESISTEC™: A crush and extraction resistant thermallycompressed tablet INTAC®: A crush and extraction resistant melt-extruded matrix SentryBond™: A crush and extraction resistant controlledrelease formulation Roxybond INTAC®: A crush and extraction resistant melt-extruded matrix Acura’s Aversion®: Extraction resistant and nasal irritating formulation RESISTEC™: A crush and extraction resistant thermally compressed tablet SentryBond™: A crush and extraction resistant formulation Suboxone Targiniq ER Troxyca ER Agonist/Antagonist: Drug-antagonist matrix Agonist/Antagonist: Drug-antagonist matrix Agonist/Antagonist: A sequestered antagonist Vantrela ER OraGuard™: A crush and extraction resistant controlled-release matrix Xartemis XR Depomed’s Acuform and Mallinckrodt’s Technology: Extraction resistant formulation DETERx®: A crush and extraction resistant controlled-release matrix OxyContin Xtampza ER Zohydro ER BeadTek™: Extraction resistant matrix Main deterring agent(s) PEO* ADF labeling Yes Naltrexone HCl Yes Cellulose acetate PEO* PEO* PEO* Acrylate copolymer Alginic acid PEO* PEO* SLS PEO* Acrylate copolymer Alginic acid Naloxone HCl Naloxone HCl Naltrexone HCl FDA status (deterred route(s)) IV Marketing status Available Available No IV upon crushing, IN, and oral upon crushing _____ Yes IV, IN, and oral upon chewing Available No Yes IV and IN No No Available _____ Available Available _____ _____ Withdrawn Available Yes IV and IN Available Yes IV and IN No Yes Yes _____ IV and IN IV upon crushing, IN, and oral upon crushing IV, IN, and oral Approved, not marketed Available Discontinued Discontinued Glyceryl behenate■ HPMC PEO* Yes Beeswax¤ Carnauba wax¥ Myristic acid§ PEO* Yes No No _____ IV and IN Discontinued Discontinued Available _____ Available * Toxic to the endothelial cells by the IV route [108]. ■Emits irritating fumes upon thermal decomposition by smoking [15] ¤ A natural excipient with toxicity concerns due to residual environmental and apicultural contaminants [95] ¥ Emits irritating fumes upon thermal decomposition by smoking [16] § Poisonous by the IV route and emits irritating fumes upon thermal decomposition by smoking [17] There are different challenges and limitations associated with each testing category. For instance, the design of category 1 studies should anticipate the various tampering methods and conditions that abusers may use. These methods could be manual (e.g., crushing with a spoon or hammer) or mechanical (e.g., coffee grinder). A major limitation with the manual manipulation methods is the variances in the applied force among different subjects, complicating the assessment process, and questioning the credibility of the results. Moreover, these studies should consider all the possible routes of abuse that in turn increase the cost, time, and the volume of the generated data. The challenges relevant to the category 2 studies include some specific circumstances where the test product cannot be evaluated for a specific route of abuse. For instance, if the presence of an excipient prohibits the IN administration of the product, the study should exclude the assessment of the IN route. Moreover, the manipulated products utilized in the studies must be reproducible to minimize the variation in the results. This requires skilled and knowledgeable personnel capable of preparing consistent samples [20]. The category 3 studies are challenged by the sample reproducibility as well as the inability to use the crossover design in certain instances. This design is recommended for the studies that measure the subjective outcomes such as in the clinical abuse potential studies. However, when blinding the test and positive control products after manipulation is not possible, another study design should be used. For example, a manipulated crush-resistant ADF can have coarse particles, while the non-ADF control can be crushed into fine powder. In this case, since the subjects can distinguish the difference in the physical appearance between the two products, a parallel group study design should be considered. The category 4 studies face more challenges as drug abuse and the related outcomes are under-reported by the abusers. Furthermore, the change in the population between the preand post-market study periods is another issue. For instance, the centers that capture the cases of abuse, misuse, overdose, etc. (e.g., poison center, treatment center), may relocate during the study period, invalidating the reference baseline for comparison. The misclassification of the abused product and data inaccuracy (e.g., the number of the abused drugs, single or multiple routes of abuse) can further confound these studies. Moreover, controlling for the concurrent interventions that impact the outcomes is another important consideration. For example, overlooking the policies addressing the prescription opioid epidemic leads to underestimated or overestimated outcomes. Lastly, the post-market studies should involve the 1258 R. AHMAD ET AL. Table 2. The required studies for the evaluation of the branded ADFs. Study Pre-market studies Category 1 (Laboratory manipulation and extraction studies) Objective Study conditions and requirements Evaluate the deterring characteristics and capacity - In-vitro study - Employing various manipulation conditions expected to alter the drug release or enable drug administration by an alternate route (e.g., heating, cooling, agitation, grinding, solvent addition, etc.) - Use of a manipulated comparator product - Testing the dissolution of intact and manipulated products - For extraction studies: Use of intact comparator and test products - For smoking studies: Use of pure active ingredient comparator, in salt and base forms Category 2 Determine the PK parameters of the manipulated - In-vivo study (Pharmacokinetic formulation - Employing the manipulation method(s) that resulted in studies) the maximum drug release in Category 1 studies - Use of intact test product and intact and manipulated comparators - Studying the food and alcohol effects - Evaluating one or more routes of administration relevant to the test product - Sampling at different points to cover the onset, peak, and offset of the effects - For the oral route: The participants are healthy volunteers who are under naltrexone blockage - For the nasal route: The participants are subjects with history of nasal abuse Category 3 (Clinical Evaluate the clinical impact - In-vivo study abuse potential - Applying a randomized, double-blind, crossover, studies) placebo- and positive-controlled study design is preferred - Employing the manipulation method(s) that resulted in the maximum drug release and plasma concentrations in Category 1 and Category 2 studies - Application of a prequalification phase; to exclude participants who cannot distinguish the placebo effect from the drug effect - Pre-qualification dose ≥lowest test dose; to exclude participants who cannot tolerate the dose of the test drug, especially for multi-dosing studies - Applying a test dose which is known to produce a high level of drug liking in recreational users - Evaluating the route of administration relevant to the test product - The participants are recreational drug users experienced with the route of abuse under evaluation - For agonist/antagonist combination products: The participants may include physically dependent population to evaluate the antagonist effect in precipitating withdrawal symptoms - Use of Visual Analogue Scales (VAS) for the assessment, bipolar or unipolar Post-market studies Category 4 Formal Determine if meaningful reductions in abuse, - Existence of a study hypothesis studies misuse, addiction, overdosing, and death have - Conduction of multiple studies by referring to different been achieved in real-world post-approval data sources to encompass the interrelated behavioral, settings clinical, and societal factors contributing to drug abuse - Full understanding of the data sources - Use of multiple branded and generic comparators - The participants are the general population with at least one study involving high-risk population while not being restricted to this specific population only - Running the study for sufficient duration - Compliance with good epidemiological practice - Validation of self-reported assessments - Considering both population and drug utilization bases to estimate the outcomes - Considering the variables (e.g., demographic and geographic) that may affect the results Category 4 Provide additional data on abuse and abuse- Use of data sources that capture diversion, tampering Supportive deterrence to help in the interpretation of the practices, prescribing trends, and street prices of information formal studies prescription drugs - Inclusion of specific populations of interest is highly recommended Outcomes - Quantity of the opioid extracted upon manipulation - Ease of drawing the drug solution into a syringe - For agonist/antagonist products: Quantity of the opioid antagonist released upon manipulation - Maximum concentration (Cmax) - Time to Cmax (tmax) - Area under the curve (AUC0-t, AUC0-∞) - Relevant partial AUC (AUC0–30, AUC0–2) - Terminal elimination halflife (t1/2) - Rate of rise of drug concentration Primary endpoint: - Immediate drug liking Secondary endpoints: - Retrospective overall drug liking - Retaking the drug - High - Alertness - Drowsiness - Nausea - For the nasal route; Intranasal irritation, runny nose, facial pain, nasal congestion Profile of subjective effects: - Onset of effect - Rate of rise to reach onset - Peak duration - Offset of effect - Abuse (frequency and prevalence) - Misuse (frequency and prevalence) - Addiction - Overdose - Death - Abuse Misuse Addiction Overdose Death EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY Table 3. Principles for the comparative evaluation of the generic products to their referenced ADFs. Principle Tier-based approach Requirement Gradual increase in the complexity of the manipulation conditions with the subsequent tiers PerformanceComprehensive knowledge based of the route of abuse assessment under evaluation and its appropriate measures Comprehensive knowledge of the abuse deterrence of the referenced ADF for the route under evaluation Most effective Tampering with the product manipulation under different thermal, mechanical, physical, and chemical conditions, and identifying all the parameters of manipulation Sample selection As a minimum, the intact drug product and the most effectively manipulated one Comparative Validation of the reference extractability point; studies <50% of the drug can be extracted from the most effectively manipulated referenced ADF within 30 minutes in any tested solvent and thermal condition Studies with Oral: Bioavailability respect to all assessment upon probable routes ingesting chewed or of abuse physically manipulated products Parenteral: Extractability and syringeability studies for both intact and manipulated products Nasal: Bioavailability and pharmacodynamic studies of sniffed manipulated products Inhalation: Sublimation potential studies on intact and manipulated products Rational Ascertain testing efficiency Avoid unnecessary steps Compare products with different formulation designs 1259 products in the oral PK studies. These studies are conducted to ensure a proper sequestration and minimal absorption of the antagonist. Another category of products with special recommendations is those incorporating aversive agents other than the ones in the referenced ADFs. In this case, neither in-vitro testing nor PK studies are feasible options. The pharmacodynamic studies that determine the clinical abuse potential (e.g., desire to retake the drug) could alternatively be used. 3. FDA-approved ADFs Maximize the extraction of the abusable agent Assess the two extreme forms Evaluate the abuse deterrence equivalency between the tested generic product and the referenced branded ADF Assess the ruggedness of the products toward all possibilities of abuse surveillance of multiple comparators, and the results should be adjusted for the population and prescription variables [21]. This requires an increased workload and time for collecting and analyzing the data. 2.2. The FDA guidance for the generic ADFs Generic opioids need to be safe and effective and display comparable abuse deterrence to the branded ADFs. The guidance, issued in 2016, and finalized in November 2017, is pertinent to solid oral opioid generics. It describes the general basics for demonstrating a deterrence equivalency between the generic products and their referenced ADFs (Table 3). The guidance provides some specific recommendations with regards to particular types of ADFs. For instance, in agonist/antagonist combinations, both ingredients must be quantified in the oral and nasal PK studies using manipulated products. Such ADFs also require an evaluation of the intact 3.1. Agonist/antagonist combination 3.1.1. Embeda In 2009, the FDA approved an ADF that could interfere with the pharmacological effect of morphine sulfate if the product is manipulated. Embeda extended-release capsules (Pfizer Inc.) is composed of multilayer pellets, where each pellet contains morphine sulfate and sequestered naltrexone HCl at a ratio of 25:1. The drug release profile in Embeda is controlled by hydrophobic polymers (ethyl cellulose and methacrylic acid copolymer) that coat the morphine sulfate layer. The sodium chloride underneath the drug layer also contributes to the drug release due to its osmotic activity [22]. Crushing the pellets is associated with an immediate release of morphine and naltrexone from the dosage form. The antagonist would, in turn, block the μ-opioid receptors upon an IN or IV administration. On the other hand, the oral administration of the intact capsules results in an adequate morphine release and low plasma concentrations of the sequestered naltrexone (if any). This assures the absence of the antagonistic effect when the medication is used as intended [23–25]. The integrity of the naltrexone sequestering coat after administering the crushed and intact Embeda capsules was evaluated in-vivo. The plasma levels of 6-β-naltrexol (naltrexone metabolite) were measured because the metabolite is more bioavailable. The results assured the rigidity of the polymeric coat upon the regular use of the product [26]. The study also revealed that the oral administration of the intact and crushed Embeda, in comparison with morphine sulfate solution, were associated with significantly lower mean scores for drug liking and drug high effects. These outcomes were attributed to the controlled-release delivery system of morphine and the release of naltrexone, respectively [26]. In another study, the oral abuse potential of morphine was compared between crushed Embeda and crushed ER morphine. The crushed Embeda, due to naltrexone release, showed statistically significant lower mean and median scores for the abuse potential responses [27]. A study by Setnik et al. [28], examined the IN abuse potential of crushed Embeda versus crushed non-ADF ER morphine. Despite having lower abuse potential measures with the crushed Embeda, 22–30% of the subjects in this study showed no reduction in the drug liking and drug high. In another study examining the IV abuse potential, the results indicated significantly lower drug high, drug liking, and euphoria for morphine/naltrexone combination or placebo than morphine alone [29]. It was reported that morphine dose dumping (up to 5 fold Cmax) is probable if co-ingested with 40% alcohol, while the naltrexone 1260 R. AHMAD ET AL. core remains intact [30]. Furthermore, the FDA determined that the oral abuse of the crushed beads is possible due to the fact that it was accompanied by euphorigenic effects [30]. The safety review of Embeda during its first year of marketing revealed that the most frequent adverse events were headache, nausea, pruritis, drug ineffectiveness, and drug withdrawal syndrome [31]. While the first three are the expected adverse effects of morphine, the drug ineffectiveness and withdrawal syndrome could be due to either the lower high experienced by the abusers with these formulations or an inferior performance of the sequestering coat and the subsequent release of naltrexone [32]. The published post-marketing data of Embeda included a case report by Ruan et al. [33], where a 50-year-old man suffering from severe opioid withdrawal symptoms was brought to the hospital. It was shown that the man crushed the product for oral abuse. Another case of withdrawal symptoms associated with the oral abuse of Embeda was reported by Jang et al. [34]. 3.1.2. Targiniq ER In September 2013, Purdue Pharma, L.P. submitted an NDA for oxycodone HCl/naloxone HCl extended-release tablets. The product was approved by the FDA in July 2014. Unlike Embeda, the antagonist in Targiniq is not sequestered and the abuse-deterrent property of the product relies on the physical and chemical difficulty of tampering with the formulation to separate the two drug molecules from each other [35]. Thus, abusing Targiniq by the IN or IV route is associated with an antagonistic effect exerted by naloxone. Characterized by its low oral bioavailability (≤2%), naloxone does not interfere with the legitimate oral drug use [36]. The product itself has no claims on oral drug abuse due to the fact that the systemic naloxone concentration is clinically insignificant to block the agonist’s effect if a chewed or crushed tablet is ingested [37]. Targiniq was available in three strengths, 5 mg/10 mg, 10 mg/20 mg, and 20 mg/40 mg (naloxone/oxycodone). Due to an intolerance to higher doses of naloxone especially in the fed state where the opioid withdrawal symptoms may appear [37], the product was never marketed at higher strengths. Targiniq ER is currently discontinued. 3.1.3. Troxyca ER Developed by Pfizer, Troxyca is an extended-release capsule consisting of oxycodone HCl and naltrexone HCl pellets. The product has been discontinued despite the fact it was approved by the FDA in 2016. The deterrence mechanism utilizes an agonist/antagonist approach [38] where naltrexone is sequestered in the pellet core. However, the sequestering coat can be disrupted if the pellets are crushed or dissolved in the selected organic solvents. This results in naltrexone release at concentrations sufficient to limit the euphoric effect of oxycodone [39]. On the other hand, the abuse-deterrent antagonist remains sequestered and preserves the therapeutic effectiveness of oxycodone if the product is used as intended. A study by Setnik et al. [40] reported lower high and drug liking scores upon an oral abuse of crushed Troxyca compared to crushed oxycodone IR formulation at similar doses (P < 0.0001). Since the bioavailability of Troxyca and the referenced non-ADF product was comparable, it was proposed that the lower scores are due to the antagonist effect of naltrexone released after the pellets were crushed [39]. In another study [41], Setnik et al. investigated the IN abuse potential of the crushed Troxyca pellets, and they observed lower scores with the ADF compared to oxycodone IR and placebo (P < 0.0001). The relative IV abuse potential of oxycodone HCl combined with naltrexone HCl (mimicking the IV administration of Troxyca ER) was evaluated by Backonja et al. [42]. The results suggested that Troxyca ER can deter the IV drug abuse if the capsule contents are crushed. In assessing the alcohol-induced dose dumping, the study showed no change in the Cmax value when the drug was administered with 20% ethanol. However, coingestion with 40% ethanol was associated with a 37% increase in the Cmax, suggesting a potentially fatal overdose if Troxyca ER is co-administered with high concentrations of alcohol [43]. 3.2. Physical/chemical barriers 3.2.1. OxyContin Oxycodone HCl tablets of Purdue Pharma L.P. was formulated as a controlled-release dosage form and approved by the FDA in 1995 [44]. The product, designed to gradually release the active agent over time with a slower onset and lower Cmax, was thought to reduce addiction. However, abusers could effectively tamper with the controlled-release formulation. In an attempt to reduce the drug abuse, Purdue reformulated the extended-release tablets late in 2007. The new formulation contained thermally cured high molecular weight poly(ethylene oxide) (PEO) [45], and the FDA approved the product in 2010. The first FDA-approved ADF resists crushing/chewing using standard methods of manipulation. Further, the product impedes syringing/injecting using aqueous solvents and resists dose dumping upon co-ingestion with alcohol [46]. Despite all the improvements, the reformulated product was still vulnerable to crushing and extraction using unconventional tampering methods. A placebo-controlled study evaluated the pharmacokinetics and pharmacodynamics of crushed reformulated tablets administered intranasally. The original OxyContin and oxycodone active ingredient were used as comparators. The results revealed lower Cmax, longer tmax and reduced overall drug liking with the reformulated OxyContin [47]. The reformulated OxyContin was found to be more effective in decreasing the drug abuse via the non-oral routes [48]. Several post-marketing data that assess the rates of abuse, overdose, and death indicated reductions in all the outcomes with the reformulated OxyContin in comparison with different extended-release (ER) and immediate-release (IR) comparators (ER morphine, ER oxymorphone, IR oxycodone, and IR hydromorphone) [48–50]. A study by Coplan et al. [51], utilizing The National Poison Data System covering all US poison centers, revealed that the abuse, therapeutic errors, and accidental exposures to reformulated OxyContin have been decreased, while increased with the other oxycodone products and heroin. Other post-marketing data confirmed that the abusers shifted to heroin and other prescription drugs, and switched to the oral abuse [52]. On the other hand, a study by Severtson et al. [53] EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY showed a decline of abuse via both oral and non-oral routes. Different studies reported that the reformulated product was associated with reduced sales and lower street prices [54,55]. Several studies conducted in Australia indicated a decline in dispensing OxyContin, especially the higher strengths [56]. Although this significantly reduced the IV abuse of the OxyContin product, it happened at the expense of greater abuse for morphine and fentanyl [56–58]. Despite the reduction in these outcomes, the drug product could still be abused by various manipulating methods. Three cases of OxyContin abuse were reported in Australia; one involved a 29-year-old female who tampered with the new formulation by breaking the tablets with kitchen shears and soaking the small parts in water and heating them. The person was admitted to the hospital several times for thrombotic microangiopathy (TMA), visual disturbances, as well as kidney function deterioration [59]. The TMA associated with the IV abuse of the product has also been reported in other studies [60,61]. It was suggested that the PEO incorporated in the new formulation has caused toxicity on the endothelial cells, resulting in TMA [59]. Different patterns of abuse for the reformulated OxyContin product have been determined by Vosburg et al. [62]. The posts on the social media and blogs reveal that the abusers have either switched to non-ADF IR products, continued the abuse of OxyContin (with microwaving, freezing before crushing, or heating in the oven), or chewed or sucked tablets orally. The post-marketing data indicated several cases of suffocation with the tablets due to the formation of a gooey mass in the abuser’s throat as well as rare incidences of intestinal obstruction that forced medical intervention [63]. 3.2.2. Hysingla ER Hysingla ER is an extended-release oral formulation of hydrocodone bitartrate that utilizes the ResistecTM Technology. The FDA approved the product in 2014 with oral, IN, and IV abuse-deterrent labeling [64]. Due to the hydrogelation of PEO, the product resists extraction in water and cannot be injected for abuse. Moreover, the in-vitro studies showed that crushing and grinding the Hysingla ER was more difficult than OxyContin [45]. On the other hand, it was found that the crushed Hysingla ER significantly loses its controlled-release feature if shaken vigorously in various heated solvents [64]. Moreover, the drug could be extracted from its formulation as a free base using organic solvents in which the deterring agent (PEO) is not soluble. Studies conducted to evaluate the abuse potential of the product showed lower Cmax and higher tmax values with the milled, chewed, and intact Hysingla ER compared to a hydrocodone oral solution [65]. Significantly lower drug liking and take drug again scores were also reported [64,66]. In another IN pharmacokinetics and abuse potential studies, the Hysingla ER product was manipulated manually and mechanically. The manipulated test product was then compared to hydrocodone powder, and the results were in favor of the ADF [67]. The safety studies reported several cases of choking and esophageal obstruction as appeared to be 1261 caused by the glue-like mass attached to the esophagus [64,68]. These studies have also shown that Hysingla ER is generally well-tolerated and can provide a 24 h. pain relief if administered as intended [69,70]. 3.2.3. Morphabond ER The extended-release product is the first morphine singleentity ADF marketed in the US [71]. It was approved in 2015 with IN and IV abuse-deterrent labeling [72]. Unlike Embeda that relies on the pharmacological effect to deter drug abuse, the SentryBondTM Technology utilized in this product offers deterrence to physical and chemical manipulations [73]. The tablet is composed of an outermost layer (diffusion layer) consisting of a homogeneously distributed morphine within ethyl acrylate and methyl methacrylate copolymer dispersion. The diffusion layer in its matrix form provides a controlled drug release even if the tablet is manipulated. This outer layer is bound to an inner acrylic copolymer barrier layer. The barrier layer improves the mechanical strength of the dosage form, providing crush resistance. The two layers are inseparable during physical manipulation of the product. This prevents any extra drug release from the inner side of the diffusion layer even after the product is crushed. An optional expansion layer is described in the US Patent 7,955,619 [74]. This third layer is covered by the insoluble barrier layer and composed of polymers (e.g., alginic acid) that expand in contact with solvents. If Morphabond ER is taken intact as prescribed, the insoluble barrier layer protects the expansion layer. Therefore, the drug release is only controlled by the diffusion layer. However, when physically compromised for subsequent IV abuse or coingestion with alcohol, the expanding polymer of the product is mixed with the diffusion-barrier layer fragments and swells in contact with solvents. This prevents drug abuse by slowing down the rate of the drug release [74]. The in-vitro syringeability and extraction volume studies confirmed the deterrence capacity of the product to an IV abuse. The interference of alcohol co-ingestion with the drug release was also excluded. The IN pharmacokinetic and dynamic assessments of crushed Morphabond ER demonstrated approximately 50% lower Cmax compared to MS Contin (morphine sulfate ER tablet) as well as lower scores of drug retaking, liking, and high [75,76]. In comparison with the oral intact Morphabond ER, similar scores and bioavailability results were obtained, indicating that the extended-release characteristics were maintained even after the dosage form was crushed [77]. The safety of the product upon its chronic use is still uncertain; the FDA has requested more post-marketing data to identify the risk of cancer, focal myocarditis, hepatotoxicity, and teratogenicity as potential outcomes of the chronic exposure to new excipients used in Morphabond ER [78]. 3.2.4. Arymo ER Arymo ER, an extended-release tablet of morphine sulfate manufactured by Egalet, was approved by the FDA in 2017. Therefore, this product is among the most recent products acquiring the ADF labeling for the IV route of abuse. The abuse deterrence by the IN route was not granted due to three years exclusivity of Morphabond that expires in October 2018 [71]. 1262 R. AHMAD ET AL. Arymo ER utilizes the Egalet’s GuardianTM Technology, where the matrix of morphine sulfate and PEO is injectionmolded under heat and high pressure [71,79,80]. This technology provided the tablets with a hardness ≥400 N, compared to 63 N for MS Contin. Furthermore, the GuardianTM Technology could maintain some of the controlled-release properties of the drug even after it was physically tampered [80]. The in-vitro manipulation by various crushing tools excluding knife and electric grinders resulted in particles larger than 500 µm. The chemical manipulation in aqueous solvents for drug extraction resulted in the formation of a viscous mass that could hardly be syringed. The studies revealed no alcohol dose dumping effect with the intact tablets. The in-vitro tests also showed that the product could not be abused by vaporization followed by smoking [81]. An oral pharmacokinetic randomized crossover study showed that the Cmax of a crushed Arymo ER is higher than that of the intact tablet and lower than that of a crushed nonADF morphine sulfate ER tablet. The oral abuse potential studies, despite showing a statistical difference in the drug liking scores between the manipulated Arymo ER and the regular extended-release morphine sulfate tablets, failed to demonstrate a statistically significant difference in the take drug again scores. Thus, the Arymo ER can still be abused orally [82]. Due to the formation of a sticky mass, choking or spitting may occur upon licking the tablet before oral administration. Additionally, swelling of the dosage form may cause intestinal obstruction, especially in patients with small gastrointestinal lumen [82]. According to the FDA, Arymo ER was found to be bioequivalent to MS Contin and it was exempted from conducting the efficacy and safety studies [83]. Egalet is still required to fully characterize the toxicity of PEO as a postmarketing requirement [81]. 3.2.5. Roxybond Roxybond, an oxycodone HCl tablet product, was approved by the FDA in 2017. It is the first immediate-release formulation labeled with abuse-deterrent properties, compliant with the FDA’s 2015 guidance for the ADFs’ evaluation and labeling [18]. Roxybond is manufactured using a similar technology employed in the other Inspiron’s ADF product, Morphabond. It consists of three functional layers; expansion, barrier, and diffusion layers as previously described with Morphabond. The SentryBond™ technology offers abuse-deterrent characteristics via the physical and chemical barriers without using antagonists or aversive agents [84]. The pre-marketing in-vitro studies conducted by Inspiron Delivery Sciences show that Roxybond resists cutting, crushing, grinding, and breaking. Roxybond can resist physical manipulation by selected household and laboratory tools, and the crushed particles are not suitable for nasal insufflation. It was concluded that Roxybond can impede drug abuse by routes that require size reduction [85]. Furthermore, both intact and manipulated forms of the drug are claimed to be resistant to solvent extraction. The formulation forms a viscous gel, resisting the passage through a needle and making the solution preparation for the IV administration more difficult [84]. The pre-marketing IN pharmacokinetics studies of the product showed lower Cmax and longer tmax when Roxybond was crushed and insufflated compared to crushed and insufflated oxycodone IR tablets, as well as intact oral Roxybond [84]. Using a bipolar Visual Analog Scale, lower overall drug liking scores of 63.84, 80.78, and 78.16 were obtained for the crushed IN Roxybond, crushed IN IR oxycodone, and intact oral Roxybond, respectively. The take drug again also followed a similar trend with 61.83, 82.07, and 76.72 scores, respectively [84,86]. 3.3. Delivery system 3.3.1. Xtampza ER Given sufficient IN and IV abuse-deterrent evidences, the extended-release oxycodone capsule product of Collegium Pharmaceutical Inc. was granted an ADF labeling in 2016. The pharmacokinetic studies showed that the crushed or chewed product could resist oral dose dumping. However, Xtampza ER score for take drug again was found to be inadequate for deterring abuse via an oral route [87]. Co-ingesting the product with different concentrations of hydroalcoholic solutions, up to 40%, as well as concomitant administration of food, showed an unlikely dose dumping effect in in-vitro and in-vivo dose dumping studies [88]. In their IN abuse potential studies, Webster et al. [89] showed that the crushed Xtampza ER is associated with lower Cmax, comparable tmax, and lower drug liking compared to the orally administered intact product. In an in-vitro study, the IV abuse potential of the product was evaluated against an extended- and immediate-release oxycodone under different conditions of abuse. Different outcomes including the percent of drug extraction, syringeability, and injectability were measured. The results showed that less than 15% of the drug could be extracted with Xtampza ER under the most challenging conditions of abuse used in the study [90]. Gudin et al. [91] compared Xtampza ER to the reformulated OxyContin. When manipulated for oral abuse, the Xtampza ER showed a greater deterrence based on its pharmacokinetic data. The Xtampza ER capsules are filled with microspheres containing waxy excipients [88], which constitute a lipid-based matrix of beeswax, carnauba wax, and myristic acid. Oxycodone in its base form is mixed with the hydrophobic matrix [92,93]. Increasing the lipophilicity of the drug substance along with the hydrophobic nature of the matrix limits the drug solubility in aqueous media. Furthermore, the waxy materials in the formulation tend to speckle rather than breaking into smaller particles upon crushing, limiting the particles’ comminution to a smaller size when the product is crushed [94]. The FDA has requested a thorough analysis of the beeswax as a natural excipient with potential residues of environmental and apicultural contaminants, along with validated test methods for detecting and quantifying such residuals [95]. Moreover, despite that the maximum permitted daily intake of oxycodone is 1.5 g, if the daily intakes of oxycodone from Xtampza ER are higher than 288 mg, the novel excipients in the drug product are required to be tested for their general toxicity, teratogenicity, effect on fertility, and carcinogenicity [95,96]. The efficacy and safety of Xtampza ER, as well as the patients’ compliance, were confirmed in patients with chronic low back pain [97]. Xtampza ER microspheres have shown to EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY be convenient for patients with swallowing difficulties; the drug substance maintains its extended-release mode from the microspheres upon mixing with soft foods, administration by nasogastric or gastrostomy feeding tubes, or chewing [88,98,99]. On the other hand, despite the efforts to overcome the food-effect on Xtampza ER, the drug bioavailability was shown to be dependent on the food intake [100]. The drug plasma levels fluctuated with the type, amount, and timing of the food intake. Moreover, with high-fat meal intake, overdosing remains to be a safety concern [95]. 3.3.2. Vantrela ER The hydrocodone bitartrate product manufactured by Teva Pharmaceuticals was approved by the FDA in 2017 after it was agreed that the product, at the minimum, showed a moderate abuse reduction via the oral, IN, and IV routes. The product is an extended-release tablet of hydrocodone granules coated with a controlled-release polymer and mixed with a gel-forming excipient [101]. The hydrophobic glyceryl behenate in the coating provides a sustained drug release. Further, it confers crush and aqueous extraction resistance properties [102]. Ethyl cellulose (EC) is also expected to provide some mechanical strength to the coating [102]. The matrix of Vantrela ER is capable of partially maintaining the extended-release pattern of hydrocodone if the drug product is crushed or dissolved in various solvents. Furthermore, due to its hydroxypropyl methylcellulose (HPMC) content, the formulation becomes too viscous in small extraction volumes, deterring the IV abuse. The in-vitro and in-vivo testing of the product in different concentrations of alcohol showed no dose-dumping [101,103]. On the other hand, pre-heating the tablet resulted in a faster drug release in some cases [101]. The product was not marketed and has recently been discontinued just one year after it was approved. 4. Drug products with abuse-deterrent features 4.1. Opana ER The oral extended-release oxymorphone tablet by Endo Pharmaceuticals approved in 2006 was first designed without abuse-deterrent features. In 2010, Endo Pharmaceuticals partnered with Grünenthal GmbH and reformulated Opana ER using a thermally extruded drug-PEO matrix. Specific extruder and processing parameters were used in the development of the product [104–106]. The new formulation intended to provide hardness and gelling properties to the tablet, making the tablet crush and extraction resistant, respectively [107]. The reformulated tablets were approved by the FDA in December 2011. However, the product was not granted the abuse-deterrent labeling due to insufficient supporting data. In early 2012, the reformulated Opana ER started replacing the original formulation in the market. In the following year, Endo submitted a supplemental application with preliminary post-marketing data, requested the FDA to approve the reformulated product as an ADF. But the FDA dismissed the request due to indecisive post-marketing data available at that time. Moreover, some data suggested the easiness of abuse via 1263 injection; 79% of the API was extractable in 5 mL solvent from thermally treated reformulated tablets [108]. Three years later, the manufacturer resubmitted the supplemental application with epidemiological data. The FDA review and discussion of the supplement was canceled following its withdrawal by Endo. Nevertheless, the preliminary review before the withdrawal revealed remarkable shifting from the IN to the IV route of abuse after the product was reformulated [109,110]. Since its launch, several cases of acute kidney injury and TMA have been reported in individuals abusing the reformulated Opana ER by injection [108, 111–113]. The Center for Biologic Evaluation and Research concluded from a study that the cause of TMA is the high molecular weight PEO (~7000,000 Da) [108]. Moreover, a study by Hunt et al. [114] showed the blockade of a dialysis catheter and tubing with a gelatinous material presented in a patient’s plasma who abused the Opana ER intravenously. The post-marketing data have also indicated an HIV outbreak in Indiana as a consequence of injecting the melted Opana ER tablets [114]. The reformulated tablets were also associated with several cases of hepatitis infection [108]. The FDA review highlighted the fact that there were limitations for the accurate interpretation of these post-marketing results. The original Opana ER was reported to be abused years after it was removed from the market, indicating the misidentification of the abused drug product. The underreporting of the overdose cases and abuse is another factor affected the accuracy of the post-marketing data. Lastly, the choice of the geographic regions covered in the studies is an additional limitation evidenced by the higher prescribing and abuse rates of Opana ER in Tennessee versus other regions. Overall, and based on the low benefit to risk ratio suggested by the post-marketing data, the FDA requested Endo Pharmaceuticals to withdraw the reformulated Opana ER from the market, and Endo voluntarily recalled the product in July 2017 [109]. 4.2. Suboxone Reckitt Benckiser Pharmaceuticals has developed sublingual tablets of buprenorphine and naloxone that was approved by the FDA in 2002 [115]. The applicant reformulated the product as sublingual films, which was later approved by the FDA in 2010. The sublingual films are less vulnerable to damage during shipping and storage and more difficult to counterfeit compared to tablets. The primary active ingredient in Suboxone is buprenorphine; a μ-opioid receptor partial agonist and a kappa receptor antagonist. This drug substance is a schedule III-controlled substance, which is prone to abuse like the other opioids. However, the approach employed in Suboxone to minimize its abuse relies on the pharmacodynamic properties of buprenorphine. As a partial agonist, the drug is associated with less euphoria and subsequently lower drug dependence potential. Additionally, the formulation incorporates an antagonist (naloxone), which in turn opposes the agonist’s effect if the product is abused by injection. The 4:1 ratio of buprenorphine to naloxone was shown to provide the maximum antagonism effect [116]. 1264 R. AHMAD ET AL. The product is intended for sublingual and buccal use, where naloxone is not absorbed at a clinically significant level [117]. The suboxone sublingual films are composed of a buffered polymeric matrix. The buffer system maintains an optimum pH of 2–4 at which the absorption of buprenorphine is maximized, while that of naloxone is minimized. It was found that the intramuscular injection of buprenorphine/naloxone was associated with the antagonistic effect, while the sublingual administration was devoid of such effect. This suggests that the product can deter abuse by injection. However, the product’s labeling indicates that Suboxone’s abuse by the IV or IN routes is possible by those having a low level of dependence to full μ-opioids or those who are dependent on partial agonists (e.g., buprenorphine) [116]. The post-marketing reports have shown hepatic abnormalities with the use of buprenorphine in general. The reports also included several cases of death as a result of overdosing by opioid-naive individuals. With regards to Suboxone, in particular, the most commonly reported post-marketing adverse events include stomatitis, glossitis, and ulceration of the mouth or tongue [116]. 4.3. Exalgo Mallinckrodt Pharmaceuticals received the FDA approval for its extended-release hydromorphone HCl product in 2010. The OROS technology (Osmotic Release Oral System, ALZA Corporation) used in the manufacturing of Exalgo exhibits some abuse-deterrent properties. Originally, the technology was developed to provide a controlled drug release over 24 h utilizing the osmosis concept. The system is composed of a semi-permeable membrane (permeable to water but not to the drug) of cellulose acetate surrounding the tablet and an inner matrix. The inner core has two layers; a drug layer containing hydromorphone HCl and a push layer containing an expandable osmopolymer (e.g., poly(ethylene oxide)) as well as an osmotic compound (e.g., sodium chloride). As water enters the inner matrix through the semipermeable membrane, the osmopolymer expands and subsequently pushes the dissolved or suspended drug out through a laser-drilled orifice at one side of the tablet’s outer membrane [118,119]. The abuse-deterrent features were suggested to be due to the rigidity of the outer cellulose acetate membrane that provides resistance to breaking and grinding, resulting in large particles that are unfit for snorting. Moreover, the fragments generate viscosity in aqueous media, making the extraction, and syringing processes harder. Sathyan et al. studied the effect of alcohol on the pharmacokinetics of Exalgo [119]. The study showed that the controlled-release properties of Exalgo tablets were maintained when co-ingested with alcohol. The bioavailability of the OROS formulation in the elderly was also reported to be marginally affected by alcohol [120]. In another study by Pande et al. [121], the product withstood physicochemical manipulations and it was to some degree difficult to abuse in comparison with a controlled-release oxycodone comparator. The post-marketing surveys of NAVIPPRO® (The National Addictions Vigilance Intervention and Prevention Program), ASI-MV substance abuse treatment center data and NPDS (The National Poison Data System) suggest a lower abuse liability via alternative routes of administration. Online posts and drug discussion forums using WIS® (Web Informed Services Internet monitoring program) also show a lower abuse desirability for the Exalgo product [122]. 4.4. Oxaydo Oxaydo is an immediate-release product of oxycodone HCl. It was developed by Acura Pharmaceuticals and received the FDA approval in 2011 with no abuse-deterrent labeling. There is no evidence to show that the Acura’s Aversion® technology incorporated in Oxaydo could actually reduce the abuse liability compared to an immediate-release oxycodone. However, the manufacturer claims that the product partially deters the physical and chemical manipulations that may discourage the IN abuse [123,124]. The history of the drug product dates to 2010 when the FDA rejected the drug application under the name Acurox. The Acurox formulation was originally developed using niacin as an aversive agent, which was determined by the FDA to offer insufficient abuse-deterrent properties [38]. It was proposed that the flushing aversive effect of niacin can easily be overcome by taking the drug with food or NSAIDs. Additionally, the undesirable effect can emerge at therapeutic doses as well, making the product unpleasant even if taken as prescribed [11]. The currently approved formulation utilizes gel-forming polymers (e.g., PEO) to deter drug extraction and syringing. Furthermore, it contains sodium lauryl sulfate (SLS), a skin and mucosal irritant that deters the nasal insufflation [38]. 4.5. Nucynta ER The FDA approved Janssen Pharmaceuticals’s extendedrelease Tapentadol HCl tablets in 2011. Nucynta ER was developed using the INTAC® technology (Grünenthal Group), where the drug is dispersed in a PEO matrix using a hot melt extrusion process [125]. The abuse-deterrent features of Nucynta ER are derived from tapentadol molecule itself as well as the INTAC® technology. Based on the NAVIPPRO ASI-MV surveillance system, tapentadol drug substance was found to have less abuse potential [126]. Similar findings were reported by DailyGovoni et al. [127], indicating a lower desirability for the nonmedical use of tapentadol in comparison with other opioids among recreational drug abusers. Another study by Vosburg et al. [128] also showed a lower rate of abuse with tapentadol versus other opioids. In terms of resistance against physical manipulation, syringing, and smoking, Nucynta ER abuse-deterrent properties were comparable to those of Oxycontin ER [129]. In comparison with Nucynta IR formulation, the extended-release version of the product was found to be less attractive to abuse [127]. The Nucynta ER is known to be abused mostly by swallowing the whole intact tablet, similar to what is reported for the ER opioids with abuse-deterrent labeling (62.5% versus 53.4% of abusers, respectively). Nucynta ER has lower abuse potential EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY by the IV route (< 15% of abusers), whereas other ER opioid products without abuse-deterrent labeling or any abusedeterrent features were shown to be abused via injection by the majority of the abusers (57.6% and 49.4%, respectively) [130]. Co-ingestion of the product with 40% alcohol was associated with an increase in the Cmax and the AUC, while the tmax was not affected [131]. 4.6. Xartemis XR A combination of oxycodone and acetaminophen (7.5 mg/ 325 mg) extended-release tablet product was approved by the FDA in 2014 with some abuse-deterrent features. The product manufactured by Mallinckrodt Pharmaceuticals is currently not available in the market. The dosage form was designed using Depomed’s Acuform technology that is not an abuse-deterrent technology in the first place. Acuform is a gastroretentive drug delivery system, relying on swellable polymers, which allow the tablet to reside longer in the stomach. Xartemis further utilizes PEO in a bilayer design; an immediate-release and an extended-release layers, both containing oxycodone and acetaminophen [132]. The fact that a smaller amount of oxycodone is contained in Xartemis compared to single entity extended-release formulations, makes Xartemis less attractive to abuse via multiple administrations. The acetaminophen’s hepatotoxicity at high doses also makes the product less desirable to abuse [133,134]. Eisenhauer et al. [135] reported that Xartemis ER, compared to an oxycodone/acetaminophen immediate-release formulation, better resists size reduction using different crushing and grinding tools except when a mortar and pestle is used. Preconditioning by heating, freezing or microwaving did not decrease the resistance of the product to size reduction. It was also observed that the crushed tablets resist extraction and form a pasty mixture in water. This mixture is difficult to draw in a syringe presumably due to the presence of PEO in the composition and the higher weight of the tablets [135]. It was also suggested that Xartemis XR is less prone to abuse by insufflation and injection [135,136]. Nevertheless, there are currently no long-term studies available to ascertain the true impact of such formulations on the opioid abuse [137]. 4.7. Zohydro ER Hydrocodone bitartrate extended-release capsules developed by Zogenix was first approved in 2014. Its approval created controversies among the health care providers regarding the impact of adding a potent opioid in a high dose formulation without any abuse-deterrent features. In response to the rising criticism [138], the company introduced a new formulation of Zohydro ER using BeadTek™ (Alkermes PLC [139]) technology in January 2017. The reformulated product failed to acquire the ADF labeling, and it is only considered to exhibit abusedeterrent features. The reformulated Zohydro ER capsules contain three different indistinguishable multi-particulate coated carrier beads; immediate-release beads, sustained-release beads, and inert beads. The immediate-release beads are sugar spheres coated with the drug and other excipients. The sustained-release 1265 beads are manufactured by coating the immediate-release beads with amino methacrylate copolymers. The combination of the immediate- and extended-release beads provide an extended release profile that was already present in the original formulation. However, in the reformulated capsules, the inert beads containing PEO can generate a viscous gel upon crushing and the addition of solvents that hinders drug extraction and syringing [139]. The pharmacokinetic studies evaluating the effect of food have shown no significant changes on AUC0-t and Cmax. Additionally, no substantial evidence was found for alcoholinduced dose-dumping when Zohydro ER was co-ingested with 20% alcohol (1.1-fold increase in Cmax compared to no alcohol ingestion). However, following co-administration with 40% alcohol, an increase in the absorption of hydrocodone (2.3-fold increase in Cmax) was reported. Also, there was an associated decrease in the tmax, suggesting that the Zohydro ER should not be taken with alcohol [140,141]. 5. Conclusion The health complications and the associated costs due to the use, misuse, and abuse of prescription opioids have increased over the past decade and resulted in the emergence of the opioid crisis. In response, ADFs have been developed, and invitro and in-vivo methods have been proposed by the FDA to evaluate the ADF products. Since 2009, several drug products exhibiting abuse-deterrent features have been introduced to the market, and some were granted the ADF labeling. While a few of these products utilize the drug delivery approach or the agonist/antagonist combination, most of these products utilize the physical/chemical barriers to deter the drug abuse. Interestingly, most of the approved ADFs are extended-release products and use the same deterring agent, i.e., PEO. Moreover, there is currently no generic prescription opioid product with an FDA-approved ADF labeling. Although most of the ADFs have been in the market for some years now, the available published post-marketing and safety data are not enough to draw final conclusions about their clinical impact in reducing the drug abuse. However, some safety concerns have been raised regarding the high molecular weight PEO. Moreover, one of the approved products (Opana ER) has been removed from the market after it was found that the abusers could abuse the drug by injection. 6. Expert opinion Safety and efficacy are prime FDA requirements and safeguard for achieving maximum but safe therapeutic effects. Manipulating prescription drugs including opioids can disrupt this safeguard and result in an unsafe access to opioid medications. As opposed to non-abusable dosage forms that are only used by patients, the ADFs are used by both patients and nonpatients (abusers). This requires the ADFs to be equipped with special but safe and powerful technologies. It is expected that the deterrent technologies employed in manufacturing of the ADFs work very effectively when the medication is manipulated for purposes other than therapy. 1266 R. AHMAD ET AL. Given the fact that the realm of drug abuse is vast, and abusers can always find more effective ways to abuse medications, the safety, and performance of the ADF products should be continuously monitored and evaluated. The FDA has so far helped with the epidemic by issuing two major guidance to deal with the branded and the generic ADFs. Although the guidance evaluates the ADFs under variety of manipulating conditions, the ADFs differ in their deterrent technologies and each can potentially be best abused by very different techniques and procedures. This requires the FDA to continuously revise its guidance to build a more comprehensive guideline to evaluate all ADFs under all routes of administrations. Given the complexity of abuse, variety of deterrent technologies, and multiple routes of drug administrations, the industry may end up with more and more ADFs with abusedeterrent features than with abuse-deterrent labeling. So far, the FDA granted the abuse-deterrent labeling to 10 out of 17 ADF products and 6 products have been denied the abusedeterrent labeling, and one has been withdrawn from the market. The ADF products that acquired the abuse-deterrent labeling have shown promising results in the pre-market in-vitro and in-vivo assessments. Regardless of the deterrent technology, all the products have shown a reduced abuse liability. Although it would take some years to better evaluate the impact of the ADFs in the real world, the post-marketing data on OxyContin confirms the potential of these formulations in resisting the drug abuse. The epidemiological results have in general indicated a decline in the different abuse outcomes, reduction in the product’s desirability, and shifting from the IN and IV routes of abuse to the less dangerous oral routes. Recognizing the shortcomings of the abuse-deterrent technologies, the deterring agents used in their formulations, and the regulatory guidelines are part of the evaluation process as well. This is critical for further development of safer and more robust formulations. As covered in this review, 11 out of 17 ADF products are based on the physical and chemical barrier approaches, mostly utilizing PEO as a deterring agent. The facts that most of the products are based on the same technology and even rely on the same deterring agent, would make it a lot easier for abusers to share their knowledge and experience and focus their efforts to maximize abuse. The most robust deterring mechanism so far seems to be the agonist/antagonist combination, whether the antagonist is blended with the drug matrix or sequestered. However, this deterring approach is still susceptible to abuse by the oral route. The formulations containing naloxone in a matrix form can be abused by chewing a single unit of the product. The formulations sequestering the antagonist are susceptible to abuse by orally administering multiple intact dosage units. The abuse via multiple administrations also applies to the other technologies and remains a major challenge. Another concern with the agonist/antagonist combination products is the food effect. Foods rich in fat enhance the solubility of the hydrophobic coating that sequesters the antagonist, forcing the release of the antagonist. The antagonist will then interfere with the agonist effect when the drug is used as intended. The majority of the approved ADFs and products with abusedeterrent features are extended-release dosage forms. This may shift the abuse efforts towards the immediate-release formulations that are available in the market without abuse-deterrent features. Therefore, it is worth considering monitoring the prescribing patterns and to equip the immediate-release formulations with abuse-deterrent technologies. The safety of the deterring agents incorporated in the ADF products is another source of concern. It is known that the maximum amount of an excipient that can safely be used in an oral dosage form can significantly be higher than when it is used via a non-oral route. Moreover, depending on how severe an ADF is manipulated, an excipient may lose its original identity to a more toxic side-product that may carry a lower lethal dose (LD50). Nevertheless, many relevant warnings are reported in the labels of different drug products and the FDA has issued two guidelines in this regard [142,143]. Besides the toxicities caused by the excipients, the impurities (e.g., organic impurities, inorganic impurities, and residual solvents) contained within these excipients pose another risk. The case is even worsened by taking these excipients at larger amounts and by routes other than those allowed in the FDA Inactive Ingredients Database [144]. The relative assessment of the deterrence efficacy of the different ADFs is another challenging aspect. The use of the invitro pre-market results as a base for the initial comparison is not feasible where the products utilize different deterring technologies requiring different outcomes. Moreover, the assessment involves multiple quantitative and qualitative measures (e.g., drug amount extracted, manipulation time, exerted effort, etc.), which complicate the evaluation process. For instance, a manipulated product showing a higher drug extraction may have undergone a longer tampering time and required multiple manipulation steps, rendering it less attractive to abusers in the real-world scenarios. The same applies for comparisons based on the pharmacokinetic and clinical abuse potential outcomes; abusers may spend a longer time and greater efforts to defeat a product’s deterring technology, rendering the product less desirable to abuse. Regardless of the deterrent technology and formulation, the post-marketing studies measure the same outcomes (e.g., abuse, misuse, and overdosing). These outcomes also reflect the products’ desirability among the abusers in the real-world settings. The comparisons based on the post-marketing studies seem to be the most reliable despite the associated challenges as shown in this review. The post-marketing studies are still ongoing for many ADF products, and it may take years before they can provide meaningful outcomes. The data is still lacking for most of the approved products, especially for those marketed recently. Moreover, some products including those with low prescription volume or low market supply may require longer post-marketing study. The development and evaluation of the ADFs are costly and may result in a higher price tag. Moreover, the market share of the new ADF products can also be affected by the position of the insurance companies whether they are willing to cover the new costly ADF products, as well as the readiness of the prescribers to adopt these products. To date, the EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY prescription of ADFs is still limited in comparison with the conventional non-ADF products [145]. Therefore, the future of the ADF products may be more promising if alternative cheaper but effective technologies are utilized in the manufacturing of the ADF products and Government or State policies are set in place to support such new products. Due to the fact that difficult manipulation of the ADFs may force the abusers to use illicit drugs, the post-marketing surveillance studies should consider the most frequently abused street drugs as additional comparators. Despite the ADFs’ potential in impeding the drug abuse, cheap non-tamper resistant street drugs are still readily available. Shifting to illicit drugs impose a serious risk and may magnify the opioid epidemic and its associated costs. The prescription opioid epidemic is a multifaceted issue and requires multifaceted solutions. Developing ADFs is only part of the solution, and a meaningful positive impact on the epidemic requires other initiatives including educational and awareness programs about the safe prescribing, dispensing, and administering of the ADFs. These programs should target physicians, pharmacists, patients, and their guardians. Moreover, naloxone availability, as well as laws and policies that enforce monitoring the doctor shopping and prescribing patterns, can also be influential. Funding This paper was not funded. Declaration of interest The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. Reviewer disclosures One reviewer is an employee of the RADARS System, which provides postmarketing surveillance data to pharma and to US FDA. One of the peer reviewers who engaged in our double-blind peer review process is affiliated to the same institution as the authors. The other peer reviewer has no relevant affiliations to declare. References Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers. 1. Opioid overdose: prescription opioids [Internet]. Atlanta (GA): CDC; 2017 [cited 2018 Jan 26]. Available from: https://www.cdc.gov/ drugoverdose/opioids/prescribed.html 2. Misuse of prescription drugs [Internet]. Bethesda (MD): NIDA; 2018 [cited 2018 Jul 22]. Available from: https://d14rmgtrwzf5a.cloud front.net/sites/default/files/2609-misuse-of-prescription-drugs.pdf 3. Vital Signs. Opioid painkiller prescribing [Internet]. Atlanta (GA): CDC; 2014 [cited 2018 Jul 22]. Available from: https://www.cdc. gov/vitalsigns/opioid-prescribing/ 4. What is the U.S. Opioid epidemic? [Internet]. Washington (DC): HHS; 2018 [cited 2018 Oct 10]. Available from: https://www.hhs. gov/opioids/about-the-epidemic/index.html 1267 5. Prescription opioid data [Internet]. Atlanta (GA): CDC; 2017 [cited 2018 Oct 10]. Available from: https://www.cdc.gov/drugoverdose/ data/prescribing.html 6. Abuse of prescription (Rx) drugs affects young adults most [Internet]. Bethesda (MD): NIH/NIDA; 2016 [cited 2018 Jul 22]. Available from: https://www.drugabuse.gov/related-topics/trendsstatistics/infographics/abuse-prescription-rx-drugs-affects-youngadults-most 7. Opioid overdose: prescription opioid overdose data [Internet]. Atlanta (GA): CDC; 2017 [cited 2018 Jan 26]. Available from: https://www.cdc.gov/drugoverdose/data/overdose.html 8. The opioid epidemic by the numbers [Internet]. Washington (DC): HHS; 2018 [cited 2018 Oct 10]. Available from: https://www.hhs. gov/opioids/sites/default/files/2018-01/opioids-infographic.pdf 9. Goodman B. Obama seeks to expand opioid addiction treatment [Internet]. New York (NY): Medscape, LLC; 2016 [cited 2018 Jan 26]. Available from: https://www.medscape.com/viewarticle/861170 10. Trends & statistics [Internet]. Bethesda (MD): NIH/NIDA; 2017 [cited 2018 Jul 22]. Available from: https://www.drugabuse.gov/ related-topics/trends-statistics'supplemental-references-foreconomic-costs 11. Hale ME, Moe D, Bond M, et al. Abuse-deterrent formulations of prescription opioid analgesics in the management of chronic noncancer pain. Pain Manag. 2016;6(5):497–508. 12. Brugal M, Barrio G, Fuente L, et al. Factors associated with non-fatal heroin overdose: assessing the effect of frequency and route of heroin administration. Addiction. 2002;97(3):319–327. 13. Novak S, Kral A. Comparing injection and non-injection routes of administration for heroin, methamphetamine, and cocaine uses in the United States. J Addict Dis. 2011;30(3):248–257. 14. Onyeka I, Basnet S, Beynon C, et al. Association between routes of drug administration and all-cause mortality among drug users. J Subst Use. 2016;21(6):559–565. 15. Glyceryl Behenate 3820–67–5. In: Lewis RJ, editor. Sax’s Dangerous Properties of Industrial Materials. Hoboken (NJ): John Wiley & Sons, Inc.; 2012. ** Pharmaceutical Excipient with Potential Toxicity. 16. Carnauba Wax 8015-86-9. In: Lewis RJ, editor. Sax’s dangerous properties of industrial materials. Hoboken (NJ): John Wiley & Sons, Inc.; 2012. doi:10.1002/0471701343.sdp30856 ** Pharmaceutical Excipient with Potential Toxicity. 17. Myristic Acid 544-63-8. In: Lewis RJ, editor. Sax’s dangerous properties of industrial materials. Hoboken (NJ): John Wiley & Sons, Inc; 2012. doi:10.1002/0471701343.sdp43863 ** Pharmaceutical Excipient with Potential Toxicity. 18. Abuse-deterrent opioids — evaluation and labeling guidance for industry [Internet]. Silver Spring (MD): FDA/CDER; 2015 [cited 2018 Jan 26]. Available from: https://www.fda.gov/ucm/groups/fdagovpublic/@fdagov-drugs-gen/documents/document/ucm334743.pdf * FDA Guidance Document. 19. General principles for evaluating the abuse deterrence of generic solid oral opioid drug products guidance for industry [Internet]. Silver Spring (MD): FDA/CDER; 2017 [cited 2018 Jan 26]. Available from: https://www.fda.gov/ucm/groups/fdagov-public/@fdagovdrugs-gen/documents/document/ucm492172.pdf * FDA Guidance Document. 20. Critical update: generic abuse deterrent opioids and the final FDA guidance [Internet]. Horsham (PA): DRUGSCAN; 2018. [cited 2018 Jul 20]. Available from: http://drugscan.com/Contents/Resources/ CATonewebinars.aspx 21. Evaluating the impact of abuse deterrent formulations: methodological challenges in postmarketing data. RADARS annual meeting; May 12; [Internet]. Denver (CO): RADARS; 2017 [cited 2018 Jul 20]. Available from: https://www.radars.org/system/events/RADARS% 20System%202017%20Annual%20Meeting_Staffa.pdf.tmp * RADARS Annual Meeting on the Challenges Associated with the Post-Marketing Studies. 22. Matthews F, Boehm G, Tang L, et al., inventors; Alpharma Pharmaceuticals LLC., assignee. Pharmaceutical composition. patent US 7,682,633. 2010 Mar 23. 1268 R. AHMAD ET AL. 23. Katz N, Sun S, Johnson F, et al. ALO-01 (Morphine sulfate and naltrexone hydrochloride) extended-release capsules in the treatment of chronic pain of osteoarthritis of the hip or knee: pharmacokinetics, efficacy, and safety. J Pain. 2010;11(4):303–311. 24. Johnson F, Ciric S, Boudriau S, et al. The relative bioavailability of morphine sulfate and naltrexone hydrochloride extended release capsules (EMBEDA®) and an extended release morphine sulfate capsule formulation (KADIAN®) in healthy adults under fasting conditions. Am J Ther. 2011;18(1):2–8. 25. Katz N, Hale M, Morris D, et al. Morphine sulfate and naltrexone hydrochloride extended release capsules in patients with chronic osteoarthritis pain. Postgrad Med. 2010;122(4):112–128. 26. Stauffer J, Setnik B, Sokolowska M, et al. Subjective effects and safety of whole and tampered morphine sulfate and naltrexone hydrochloride (ALO-01) extended-release capsules versus morphine solution and placebo in experienced non-dependent opioid users. A randomized, double-blind, placebo-controlled, crossover study. Clin Drug Investig. 2009;29(12):777–790. 27. EMBEDA® (morphine sulfate and naltrexone hydrochloride) extended-release capsules, for oral use, CII package insert [Internet]. New York (NY): Pfizer Inc.; 2016 [cited 2017 Jun 30]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/ label/2016/022321s022lbl.pdf 28. Setnik B, Goli V, Levy-Cooperman N, et al. Assessing the subjective and physiological effects of intranasally administered crushed extended-release morphine formulations with and without a sequestered naltrexone core in recreational opioid users. Pain Res Manag. 2013;18(4):e55–e62. 29. Webster L, Johnson F, Stauffer J, et al. Impact of intravenous naltrexone on intravenous morphine-induced high, drug liking, and euphoric effects in experienced, nondependent male opioid users. Drugs R D. 2011;11(3):259–275. 30. Application number: 22-321 summary review [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2008 [cited 2017 Dec 3]. Available from: https://www.accessdata.fda.gov/ drugsatfda_docs/nda/2009/022321s000SumR.pdf 31. Badalamenti V, Buckley J, Smith E. Safety of Embeda® (Morphine sulfate and naltrexone hydrochloride) extended-release capsules: review of postmarketing adverse events during the first year. J Opioid Manag. 2012;8(2):115–125. 32. West R, Guevara M, Mikel C. Detection of naltrexone and naltrexol in patients prescribed Embeda®. J Opioid Manag. 2017;13 (3):139–140. 33. Ruan X, Chen T, Gudin J, et al. Acute opioid withdrawal precipitated by ingestion of crushed embeda (Morphine extended release with sequestered naltrexone): case report and the focused review of the literature. J Opioid Manag. 2010;6(4):300–303. 34. Jang D, Rohe J, Hoffman R. Severe opioid withdrawal due to misuse of new combined morphine and naltrexone product (Embeda). Ann Emerg Med. 2010;55(3):303–304. 35. Caruso F, Kao H, inventors; Purdue Pharma L.P., assignee. Abuseresistant controlled-release opioid dosage form. patent US 9,511,066. 2016 Dec 6. 36. Application number: 205777Orig1s000 medical review(s) [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2014 [cited 2017 Sep 9]. Available from: https://www.accessdata. fda.gov/drugsatfda_docs/nda/2014/205777Orig1s000MedR.pdf 37. Application number: 205777Orig1s000 clinical pharmacology and biopharmaceutics review(s) [Internet]. Silver Spring (MD): FDA/ Center for Drug Evaluation and Research; 2014 [cited 2017 Jul 1]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/ nda/2014/205777Orig1s000ClinPharmR.pdf 38. Mastropietro D, Omidian H. Abuse-deterrent formulations: part 2: commercial products and proprietary technologies. Expert Opin Pharmacother. 2015;16(3):305–323. 39. FDA briefing document joint meeting of anesthetic and analgesic drug products advisory committee and drug safety and risk management advisory committee [Internet]. Silver Spring (MD): FDA; 2016 [cited 2018 Jan 29]. Available from: https://www.fda.gov/ downloads/AdvisoryCommittees/CommitteesMeetingMaterials/ 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. ** 52. 53. 54. 55. 56. 57. Drugs/AnestheticAndAnalgesicDrugProductsAdvisoryCommittee/ UCM505122.pdf Setnik B, Bass A, Bramson C, et al. Abuse potential study of ALO-02 (Extended-release oxycodone surrounding sequestered naltrexone) compared with immediate-release oxycodone administered orally to nondependent recreational opioid users. Pain Med. 2017;18 (6):1077–1088. Setnik B, Bramson C, Bass A, et al. Intranasal administration of crushed ALO-02 (extended-release oxycodone with sequestered naltrexone): a randomized, controlled abuse-potential study in nondependent recreational opioid users. J Clin Pharmacol. 2015;55(12):1351–1361. Backonja M, Webster L, Setnik B, et al. Intravenous abuse potential study of oxycodone alone or in combination with naltrexone in nondependent recreational opioid users. Am J Drug Alcohol Abuse. 2016;42(5):539–549. Malhotra B, Matschke K, Wang Q, et al. Effects of ethanol on the pharmacokinetics of extended-release oxycodone with sequestered naltrexone (ALO-02). Clin Drug Invest. 2015;35(4):267–274. Application number: 22-272 medical review(s) [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2009 [cited 2018 Jan 22]. Available from: https://www.accessdata.fda. gov/drugsatfda_docs/nda/2010/022272s000MedR.pdf McKenna W, Mannion R, O’Donnell E, et al., inventors; Purdue Pharma LP, Purdue Pharmaceuticals LP, assignee. Tamper resistant dosage forms. patent US 9,770,416. 2017 Sep 26. Application number: 22-272 clinical pharmacology and biopharmaceutics review(s) [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2009 [cited 2018 Jan 22]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2010/ 022272s000ClinPharmR.pdf Harris S, Perrino P, Smith I, et al. Abuse potential, pharmacokinetics, pharmacodynamics, and safety of intranasally administered crushed oxycodone HCl abuse-deterrent controlled-release tablets in recreational opioid users. J Clin Pharmacol. 2014;54 (4):468–477. Coplan P, Chilcoat H, Butler S, et al. The effect of an abuse-deterrent opioid formulation (OxyContin) on opioid abuse-related outcomes in the postmarketing setting. Clin Pharmacol Ther. 2016;100(3):275–286. Sessler N, Downing J, Kale H, et al. Reductions in reported deaths following the introduction of extended-release oxycodone (OxyContin) with an abuse-deterrent formulation. Pharmacoepidemiol Drug Saf. 2014;23(12):1238–1246. Butler S, Cassidy T, Chilcoat H, et al. Abuse rates and routes of administration of reformulated extended-release oxycodone: initial findings from a sentinel surveillance sample of individuals assessed for substance abuse treatment. J Pain. 2013;14(4):351–358. Coplan P, Kale H, Sandstrom L, et al. Changes in oxycodone and heroin exposures in the national poison data system after introduction of extended-release oxycodone with abuse-deterrent characteristics. Pharmacoepidemiol Drug Saf. 2013;22(12):1274–1282. Shifting to heroin abuse. Cicero T, Ellis M. Abuse-deterrent formulations and the prescription opioid abuse epidemic in the United States-lessons learned from OxyContin. JAMA Psychiatry. 2015;72(5):424–429. Severtson S, Ellis M, Kurtz S, et al. Sustained reduction of diversion and abuse after introduction of an abuse deterrent formulation of extended release oxycodone. Drug Alcohol Depend. 2016;168:219–229. Hwang C, Chang H, Alexander G. Impact of abuse-deterrent OxyContin on prescription opioid utilization. Pharmacoepidemiol Drug Saf. 2015;24(2):197–204. Severtson S, Bartelson B, Davis J, et al. Reduced abuse, therapeutic errors, and diversion following reformulation of extended-release oxycodone in 2010. J Pain. 2013;14(10):1122–1130. Schaffer A, Buckley N, Degenhardt L, et al. Person-level changes in oxycodone use after the introduction of a tamper-resistant formulation in Australia. Cmaj. 2018;190(12):E355–E362. Degenhardt L, Bruno R, Ali R, et al. The introduction of a potentially abuse deterrent oxycodone formulation: early findings from the EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 58. 59. 60. 61. 62. * 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. Australian National Opioid Medications Abuse Deterrence (NOMAD) study. Drug Alcohol Depend. 2015;151:56–67. Jauncey M, Livingston M, Salmon A, et al. The impact of OxyContin reformulation at the Sydney medically supervised injecting centre: pros and cons. Int J Drug Policy. 2018;53:17–22. Nataatmadja M, Divi D. Relapsing thrombotic microangiopathy and intravenous sustained-release oxycodone. Clin Kidney J. 2016;9 (4):580–582. Robson K, Clucas D, Filshie R, et al. Thrombotic microangiopathy associated with intravenous injection of extended-release oxycodone. BMJ Case Rep. 2017 [cited 2018 Jan 22];1–3. DOI:10.1136/bcr-2017-220977 Tate C, Mollee P. Intravenous oxycontin-associated thrombotic microangiopathy treated successfully without plasma exchange. Med J Aust. 2015;202(6):330–332. Vosburg S, Haynes C, Besharat A, et al. Changes in drug use patterns reported on the web after the introduction of ADF Oxycontin: findings from the researched abuse, diversion, and addiction-related surveillance (RADARS) system web monitoring program. Pharmacoepidemiol Drug Saf. 2017;26:1044–1052. Patterns of oxycontin abuse. OXYCONTIN® (oxycodone hydrochloride) extended-release tablets, for oral use, CII package insert [Internet]. Stamford (CT): Purdue Pharma L.P.; 2016 [cited 2017 Nov 16]. Available from: https://www. accessdata.fda.gov/drugsatfda_docs/label/2016/022272s034lbl.pdf Application number: 206627Orig1s000 summary review [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2014 [cited 2017 Nov 28]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/ 206627Orig1s000SumR.pdf Harris S, Cipriano A, Colucci S, et al. Oral abuse potential, pharmacokinetics, and safety of once-daily, single-entity, extended-release hydrocodone (HYD) in recreational opioid users. Pain Med. 2017;18:1278–1291. HYSINGLA ER (hydrocodone bitartrate) extended-release tablets, for oral use, CII package insert [Internet]. Stamford (CT): Purdue Pharma L.P.; 2014 [cited 2017 Nov 30]. Available from: https://www. accessdata.fda.gov/drugsatfda_docs/label/2016/206627s004lbl.pdf Harris S, Cipriano A, Colucci S, et al. Intranasal abuse potential, pharmacokinetics, and safety of once-daily, single-entity, extended-release hydrocodone (HYD) in recreational opioid users. Pain Med. 2016;17(5):820–831. Long-term safety of once-daily hydrocodone bitartrate (HYD) tablets for moderate to severe chronic nonmalignant and nonneuropathic pain. Includes a 24-week extension period [Internet]. Bethesda (MD): NIH/U.S. National Library of Medicine; 2017 [cited 2017 Nov 29]. Available from: https://clinicaltrials.gov/ct2/show/ results/NCT01400139?sect=X340156'evnt Kapil R, Cipriano A, Wen W, et al. Pharmacokinetic profile and sustained 24-hour analgesia of a once-daily hydrocodone bitartrate extended-release tablet with abuse-deterrent properties. Clin Ther. 2016;38(2):302–314. Taber L, Lynch S, He E, et al. Long-term safety and effectiveness of once-daily, single-entity, extended-release hydrocodone over 76 weeks of an open-label study in patients with chronic noncancer and nonneuropathic pain. Postgrad Med. 2016;128(1):23–33. Application number: 208603Orig1s000 summary review [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2017 [cited 2017 Nov 28]. Available from: https://www.accessdata. fda.gov/drugsatfda_docs/nda/2017/208603Orig1s000SumR.pdf Application number: 206544Orig1s000 summary review [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2015 [cited 2017 Dec 2]. Available from: https://www.accessdata. fda.gov/drugsatfda_docs/nda/2015/206544Orig1s000SumR.pdf Inspirion delivery technologies receives FDA approval for MorphaBond™ (morphine sulfate) extended-release tablets CII, an opioid analgesic formulated with abuse-deterrent properties [Internet]. Valley Cottage (NY): PR Newswire; 2015 [cited 2017 Dec 2]. Available from: http://www.prnewswire.com/news-releases /inspirion-delivery-technologies-receives-fda-approval-for- 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. 87. 88. 1269 morphabond-morphine-sulfate-extended-release-tablets-cii-anopioid-analgesic-formulated-with-abuse-deterrent-properties -300153910.html Shah M, Difalco R, inventors; Inspirion Delivery Technologies, LLC., assignee. Abuse resistant drugs, method of use and method of making. patent US 7,955,619. 2011 Jun 7. Application number: 206544Orig1s000 clinical pharmacology and biopharmaceutics review(s) [Internet]. Silver Spring (MD): FDA/ Center for Drug Evaluation and Research; 2015 [cited 2017 Dec 2]. Available from: https://www.accessdata.fda.gov/drugsatfda_ docs/nda/2015/206544Orig1s000ClinPharmR.pdf Webster L, Pantaleon C, Shah M, et al. A randomized, double-blind, double-dummy, placebo-controlled, intranasal drug liking study on a novel abuse-deterrent formulation of morphine—morphine ARER. Pain Med. 2017;18:1303–1313. MORPHABOND ER™ (morphine sulfate) extended-release tablets, for oral use CII Package Insert [Internet]. Basking Ridge (NJ): Inspirion Delivery Sciences, LLC; 2016 [cited 2017 Dec 2]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/ label/2016/206544s002s005lbl.pdf Application number: 206544Orig1s000 pharmacology review(s) [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2015 [cited 2017 Dec 3]. Available from: https://www. accessdata.fda.gov/drugsatfda_docs/nda/2015/ 206544Orig1s000PharmR.pdf Tygesen P, Lindhardt K, Olsen M, et al., inventors; EGALET LTD., assignee. Abuse deterrent pharmaceutical compositions for controlled release. patent US 9,549,899. 2017 Jan 24. Sponsor briefing document ARYMO™ ER (Morphine sulfate) extended-release tablets. Joint Meeting of the Anesthetic and Analgesic Drug Products Advisory Committee and the Drug Safety and Risk Management Advisory Committee [Internet]. Egalet; 2016 [cited 2017 Dec 1]. Available from: https://www.fda.gov/downloads/ AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/ AnestheticAndAnalgesicDrugProductsAdvisoryCommittee/ UCM514383.pdf Application number: 208603Orig1s000 chemistry review(s) [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2016 [cited 2017 Nov 30]. Available from: https://www. accessdata.fda.gov/drugsatfda_docs/nda/2017/ 208603Orig1s000ChemR.pdf ARYMO™ ER (morphine sulfate) extended-release tablets, for oral use CII package insert [Internet]. Wayne (PA): Egalet US Inc.; 2017 [cited 2017 Nov 29]. Available from: https://www.accessdata.fda. gov/drugsatfda_docs/label/2017/208603s000lbl.pdf Application number: 208603Orig1s000 clinical pharmacology and biopharmaceutics review(s) [Internet]. Silver Spring (MD): FDA/ Center for Drug Evaluation and Research; 2016 [cited 2017 Dec 1]. Available from: https://www.accessdata.fda.gov/drugsatfda_ docs/nda/2017/208603Orig1s000ClinPharmR.pdf Daiichi Sankyo, Inc. and inspirion delivery sciences LLC announce plans for commercialization of RoxyBond™ (oxycodone hydrochloride) Tablets CII in the U.S. [Internet]. Parsippany (NJ): Daiichi Sankyo, Inc; 2018 [cited 2018 Jan 29]. Available from: http://www. daiichisankyo.com/media_investors/media_relations/press_ releases/detail/006638.html Kinzler E, Pantaleon C, Iverson M, et al. Oxycodone ARIR (RoxyBond™) is resistant to physical manipulation techniques commonly used by opioid abusers. Postgrad Med. 2017;129(Sup1):16. Webster L, Iverson M, Pantaleon C, et al. A randomized, double-blind, double-dummy, placebo-controlled, intranasal human abuse potential study of oxycodone ARIR, a novel, immediate-release, abuse-deterrent formulation. Pain Med. 2018; pny043–pny043. DOI:10.1093/pm/pny043. Application number: 208090Orig1s000 summary review [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2016 [cited 2017 Jul 4]. Available from: https://www.accessdata.fda. gov/drugsatfda_docs/nda/2016/208090Orig1s000SumR.pdf Application number: 208090Orig1s000 chemistry review(s) [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and 1270 89. 90. 91. 92. 93. 94. 95. * 96. 97. 98. 99. 100. 101. 102. 103. 104. 105. R. AHMAD ET AL. Research; 2016 [cited 2017 Jul 3]. Available from: https://www. accessdata.fda.gov/drugsatfda_docs/nda/2016/ 208090Orig1s000ChemR.pdf Webster L, Kopecky E, Smith M, et al. A randomized, double-blind, double-dummy study to evaluate the intranasal human abuse potential and pharmacokinetics of a novel extended-release abuse-deterrent formulation of oxycodone. Pain Med. 2016;17 (6):1112–1130. Fleming A, Scungio T, Grima M, et al. In vitro assessment of the potential for abuse via the intravenous route of oxycodone DETERx® microspheres. J Opioid Manag. 2016;12(1):57–65. Gudin J, Levy-Cooperman N, Kopecky E, et al. Comparing the effect of tampering on the oral pharmacokinetic profiles of two extended-release oxycodone formulations with abuse-deterrent properties. Pain Med. 2015;16(11):2142–2151. Rariy R, Fleming A, Hirsh J, et al., inventors; Collegium Pharmaceutical Inc, assignee. Abuse-deterrent pharmaceutical compositions of opioids and other drugs. patent US 9,248,195. 2016 Feb 2. XTAMPZA ER (oxycodone) extended-release capsules, for oral use, CII Package Insert [Internet]. Canton (MA): Collegium Pharmaceutical Inc.; 2016 [cited 2017 Jul 5]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2016/ 208090s003lbl.pdf Mayock S, Saim S, Fleming A. In vitro drug release after crushing: evaluation of Xtampza ER and other ER opioid formulations. Clin Drug Invest. 2017;37:1117–1124. Application number: 208090Orig1s000 medical review(s) [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2015 [cited 2017 Jul 6]. Available from: https://www.accessdata.fda. gov/drugsatfda_docs/nda/2016/208090Orig1s000MedR.pdf Pharmaceutical Excipient with Potential Toxicity. Application number: 208090Orig1s000 pharmacology review(s) [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2015 [cited 2017 Jul 6]. Available from: https://www.access data.fda.gov/drugsatfda_docs/nda/2016/208090Orig1s000PhamR.pdf Katz N, Kopecky E, O’Connor M, et al. A phase 3, multicenter, randomized, double-blind, placebo-controlled, safety, tolerability, and efficacy study of Xtampza ER in patients with moderate-tosevere chronic low back pain. Pain. 2015;156(12):2458–2467. McCarberg B, Kopecky E, O’Connor M, et al. An abuse-deterrent, microsphere-in-capsule formulation of extended-release Oxycodone: alternative modes of administration to facilitate pain management in patients with dysphagia. Curr Med Res Opin. 2016;32(12):1975–1982. Kopecky E, Fleming A, Levy-Cooperman N, et al. Oral human abuse potential of Oxycodone DETERx® (Xtampza® ER). J Clin Pharmacol. 2017;57(4):500–512. Application number: 208090Orig1s000 clinical pharmacology and biopharmaceutics review(s) [Internet]. Silver Spring (MD): FDA/ Center for Drug Evaluation and Research; 2015 [cited 2017 Jul 5]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/ nda/2016/208090Orig1s000ClinPharmR.pdf Application number: 207975Orig1s000 chemistry review(s) [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2015 [cited 2017 Jul 2]. Available from: https://www.accessdata.fda. gov/drugsatfda_docs/nda/2017/207975Orig1s000ChemR.pdf Habib W, Hamed E, Zepeda M, inventors; Cima Labs Inc., assignee. Abuse resistant drug formulation. patent US 9,216,176. 2015 Dec 22. Application number : 207975Orig1s000 summary review [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2017 [cited 2017 Jul 2]. Available from: https://www.accessdata.fda. gov/drugsatfda_docs/nda/2017/207975Orig1s000SumR.pdf Arkenau-Maric E, Bartholomaus J, Kugelmann H, inventors; Grunenthal GmbH, assignee. Abuse-proof dosage form. patent US 8,192,722. 2012 Jun 1. Application number: 201655Orig1s000 chemistry review(s) [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2011 [cited 2018 Jan 20]. Available from: https://www. 106. 107. 108. • 109. * 110. * 111. 112. 113. 114. 115. 116. 117. 118. accessdata.fda.gov/drugsatfda_docs/nda/2011/ 201655Orig1s000ChemR.pdf Application number: 201655Orig1s000 clinical pharmacology and biopharmaceutics review(s) [Internet]. Silver Spring (MD): FDA/ Center for Drug Evaluation and Research; 2011 [cited 2018 Jan 24]. Available from: https://www.accessdata.fda.gov/drugsatfda_ docs/nda/2011/201655Orig1s000ClinPharmR.pdf Vosburg S, Jones J, Manubay J, et al. Assessment of a formulation designed to be crush-resistant in prescription opioid abusers. Drug Alcohol Depend. 2012;126:206–215. FDA Document on Safety Issues Related to the Withdrawn Opana ER: FDA briefing document. Joint Meeting of the Drug Safety and Risk Management (DSaRM) Advisory Committee and the Anesthetic and Analgesic Drug Products Advisory Committee (AADPAC) Meeting; March 13–14, 2017. Postmarketing Safety Issues Related to Reformulated Opana ER® [Internet]. Silver Spring (MD): FDA/ CDER/Office of Surveillance and Epidemiology (OSE); 2017 [cited 2017 Dec 6]. Available from: https://www.fda.gov/downloads/ AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/ AnestheticAndAnalgesicDrugProductsAdvisoryCommittee/ UCM545760.pdf FDA Document on safety issues related to the Withdrawn Opana ER. FDA postmarket drug safety information for patients and providers. Oxymorphone (marketed as Opana ER) information [Internet]. Silver Spring (MD): FDA; 2017 [cited 2017 Dec 18]. Available from: https://www.fda.gov/drugs/drugsafety/postmarketdrugsafetyinfor mationforpatientsandproviders/ucm562339.htm FDA Document on the Withdrawn Opana ER. FDA briefing document. Joint Meeting of the Drug Safety and Risk Management (DSaRM) Advisory Committee and the Anesthetic and Analgesic Drug Products Advisory Committee (AADPAC) Meeting; March 13–14, 2017. Postmarketing Safety Issues Related to Reformulated Opana ER® Addendum [Internet]. Silver Spring (MD): FDA/CDER/Office of Surveillance and Epidemiology (OSE); 2017 [cited 2017 Dec 19]. Available from: https://www.fda.gov/downloads/advi sorycommittees/committeesmeetingmaterials/drugs/anestheticanda nalgesicdrugproductsadvisorycommittee/ucm545761.pdf FDA Document on the Withdrawn Opana ER. Gandhi A, Ullah S, Kotadia S, et al. Oxymorphone induced thrombotic microangiopathy mimicking atypical haemolytic uremic syndrome. J Ayub Med Coll Abbottabad. 2016;29(2):360–362. Ambruzs J, Serrell P, Rahim N, et al. Thrombotic microangiopathy and acute kidney injury associated with intravenous abuse of an oral extended-release formulation of oxymorphone hydrochloride: kidney biopsy findings and report of 3 cases. Am J Kidney Dis. 2014;63(6):1022–1026. Kotbi N, Han B, Cheng D. Opana® ER induced thrombotic thrombocytopenic purpura. Int Med Case Rep J. 2015;8:97–98. Hunt R, Yalamanoglu A, Tumlin J, et al. A mechanistic investigation of thrombotic microangiopathy associated with IV abuse of Opana ER. Blood. 2017;129(7):896–905. Application number: 20-732 and 20-733 approval letter(s) [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2002 [cited 2018 Feb 24]. Available from: https://www.accessdata. fda.gov/drugsatfda_docs/nda/2002/20-733_Subutex_Approv.pdf SUBOXONE® (buprenorphine and naloxone) sublingual film, for sublingual or buccal use CIII package insert [Internet]. North Chesterfield (VA): Indivior Inc; 2018 [cited 2018 Feb 24]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/ 022410s031lbl.pdf Application number: 022410Orig1s000 chemistry review(s) [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2009 [cited 2018 Feb 26]. Available from: https://www.accessdata. fda.gov/drugsatfda_docs/nda/2010/022410Orig1s000ChemR.pdf Siegel RA, Rathbone MJ. Overview of controlled release mechanisms. In: Siepmann J, Siegel RA, Rathbone MJ, editors. Fundamentals and applications of controlled release drug delivery. Boston (MA): Springer US; 2012. p. 19–43. EXPERT OPINION ON DRUG METABOLISM & TOXICOLOGY 119. Sathyan G, Sivakumar K, Thipphawong J. Pharmacokinetic profile of a 24-hour controlled-release OROS formulation of hydromorphone in the presence of alcohol. Curr Med Res Opin. 2008;24(1):297–305. 120. Lussier D, Richarz U, Finco G. Use of hydromorphone, with particular reference to the OROS® formulation, in the elderly. Drugs Aging. 2010;27(4):327–335. 121. Pande P, Hines J, Brogan A. Tamper-resistant properties of once-daily hydromorphone ER (OROS hydromorphone). J Pain. 2011;12(4):P58. 122. Butler S, McNaughton E, Black R, et al. Evaluation of the relative abuse of an OROS(R) extended-release hydromorphone HCI product: results from three post-market surveillance studies. Clin J Pain. 2018;34(7):618–628. 123. OXAYDO® (oxycodone HCl) tablets for oral use only, CII, package insert [Internet]. Wayne (PA): Egalet US Inc; 2016 [cited 2018 Jan 30]. Available from: https://www.accessdata.fda.gov/drugsatfda_ docs/label/2016/202080s005s006lbl.pdf 124. How could a pain treatment affect the community? [Internet]. Wayne (PA): Egalet US Inc; 2017 [cited 2018 Jan 30]. Available from: http://www.oxaydo.com/hcp/ 125. Maincent J, Zhang F. Recent advances in abuse-deterrent technologies for the delivery of opioids. Int J Pharm. 2016;510(1):57–72. 126. Butler SF, McNaughton EC, Black RA. Tapentadol abuse potential: a postmarketing evaluation using a sample of individuals evaluated for substance abuse treatment. Pain Med. 2015;16(1):119–130. 127. Dailey-Govoni T, Beaumont J, Cassidy T. PAINWeek abstract book 2017: profiling non-medical use of tapentadol products among recreational drug abusers. Postgrad Med. 2017;129(Sup 1):76–77. 128. Vosburg S, Severtson S, Dart R, et al. Assessment of tapentadol API abuse liability with the researched abuse, diversion and addiction-related surveillance (RADARS) system. J Pain. 2017;19 (4):439–453. 129. Cone E, McGoff P, Sokolowska M, et al. PAINWeek abstract book 2017: characteristics of tapentadol extended-release formulation. Postgrad Med. 2017;129(Sup 1):44–45. 130. Beaumont J, Govoni T, Cassidy T. PAINWeek abstract book 2017: abuse profile of Nucynta ER compared to extended-release opioids with and without FDA abuse-deterrent labeling. Postgrad Med. 2017;129(Sup 1):27–28. 131. Application number: 200533Orig1s000 summary review [Internet]. Silver Spring (MD): FDA/CDER; 2011 [cited 2018 Jul 25]. Available from: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/ 200533Orig1s000SumR.pdf 132. Devarakonda K, Guiliani M, Gupta V, et al., inventors; Mallinckrodt LLC, assignee. Combination composition comprising oxycodone and acetaminophen for rapid onset and extended duration of analgesia. Patent US 8,658,631. 2014 Feb 25. 133. Morton T, Kostenbader K, Montgomery J, et al. Comparison of subjective effects of extended-release versus immediate-release oxycodone/acetaminophen tablets in healthy nondependent recreational users of prescription opioids: a randomized trial. Postgrad Med. 2014;126(4):20–32. 1271 134. Morton T, Devarakonda K, Kostenbader K, et al. Correlation of subjective effects with systemic opioid exposure from fixed-dose combinations of oxycodone/acetaminophen in recreational users of prescription drugs. Pain Med. 2016;17(3):539–550. 135. Eisenhauer T, Matchett M, Heasley R, et al. Evaluation of the tamper-resistant properties of biphasic immediate-release/ extended-release oxycodone/acetaminophen tablets. Drug Dev Ind Pharm. 2016;42(1):157–165. 136. Vadivelu N, Schermer E, Kodumudi G, et al. The clinical applications of extended-release abuse-deterrent opioids. CNS Drugs. 2016;30 (7):637–646. 137. Pergolizzi J, Taylor R, Raffa R. The potential role of an extended-release, abuse-deterrent oxycodone/acetaminophen fixed-dose combination product for the treatment of acute pain. Adv Ther. 2015;32(6):485–495. 138. Gould H, Paul D. Hydrocodone extended-release: pharmacodynamics, pharmacokinetics and behavioral pharmacology of a controversy. Pharmacol Res. 2015;91:99–103. 139. Rekhi G, Sidwell R, inventors; Alkermes Pharma Ireland Limited, assignee. Abuse resistant pharmaceutical compositions. patent US 9,132,096. 2015 Sep 15. 140. Farr SJ, Robinson CY, Rubino CM. Effects of food and alcohol on the pharmacokinetics of an oral, extended-release formulation of hydrocodone in healthy volunteers. Clin Pharmacol Adv Appl. 2015;7:1–9. 141. Giordano J, Huang H, Kianto J, et al. In vitro assessment of the effects of alcohol on the release rate of extended-release opioid formulations (Hysingla® ER, OxyContin® and Zohydro® ER). J Pain. 2016;17(4,Supplement):S65–S66. 142. Guidance for industry nonclinical studies for the safety evaluation of pharmaceutical excipients [Internet]. Silver Spring (MD): FDA/ Center for Drug Evaluation and Research, Center for Biologics Evaluation and Research; 2005 [cited 2017 Nov 20]. Available from: https://www.fda.gov/ucm/groups/fdagov-public/documents/ document/ucm079250.pdf 143. Guidance for industry: limiting the use of certain phthalates as excipients in CDER-regulated products [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2012 [cited 2018 Mar 1]. Available from: https://www.fda.gov/downloads/ Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ UCM294086.pdf 144. Inactive ingredient search for approved drug products [Internet]. Silver Spring (MD): FDA/Center for Drug Evaluation and Research; 2018 [cited 2018 Mar 1]. Available from: https://www.accessdata. fda.gov/scripts/cder/iig/index.cfm 145. Statement from FDA commissioner Scott Gottlieb, M.D., on steps to promote development of generic versions of opioids formulated to deter abuse [Internet]. Silver Spring (MD): FDA; 2018 [cited 2018 Oct 10]. Available from: https://www. fda.gov/NewsEvents/Newsroom/PressAnnouncements/ ucm586117.htm