throbber
Journal of Controlled Release 183 (2014) 154–166
`
`Contents lists available at ScienceDirect
`
`Journal of Controlled Release
`
`j o u r n a l h o m e p a g e : w w w . e l s e v i e r . c o m / l o c a t e / j c o n r e l
`
`Review
`Naltrexone: A review of existing sustained drug delivery systems and
`emerging nano-based systems
`Nowsheen Goonoo a, Archana Bhaw-Luximon a, Reetesh Ujoodha a, Anil Jhugroo b,
`Gary K. Hulse c, Dhanjay Jhurry a,⁎
`a ANDI Centre of Excellence for Biomedical and Biomaterials Research, MSIRI Building, University of Mauritius, Réduit, Mauritius
`b Dept. of Medicine, University of Mauritius, Réduit, Mauritius
`c School of Psychiatry and Clinical Neurosciences, The University of Western Australia, M521, D Block, QEII Medical Centre, Nedlands, WA 6009, Australia
`
`a r t i c l e
`
`i n f o
`
`a b s t r a c t
`
`Article history:
`Received 10 February 2014
`Accepted 24 March 2014
`Available online 2 April 2014
`
`Keywords:
`Naltrexone
`Naltrexone sustained release formulations and
`safety
`Naltrexone-loaded nanocarriers and nanogels
`
`Narcotic antagonists such as naltrexone (NTX) have shown some efficiency in the treatment of both opiate addic-
`tion and alcohol dependence. A few review articles have focused on clinical findings and pharmacogenetics of
`NTX, advantages and limitations of sustained release systems as well as pharmacological studies of NTX depot
`formulations for the treatment of alcohol and opioid dependency. To date, three NTX implant systems have
`been developed and tested in humans. In this review, we summarize the latest clinical data on commercially
`available injectable and implantable NTX-sustained release systems and discuss their safety and tolerability
`aspects. Emphasis is also laid on recent developments in the area of nanodrug delivery such as NTX-loaded
`micelles and nanogels as well as related research avenues. Due to their ability to increase the therapeutic
`index and to improve the selectivity of drugs (targeted delivery), nanodrug delivery systems are considered as
`promising sustainable drug carriers for NTX in addressing opiate and alcohol dependence.
`© 2014 Elsevier B.V. All rights reserved.
`
`Contents
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`155
`155
`155
`155
`156
`156
`157
`158
`158
`159
`159
`159
`160
`160
`161
`161
`162
`163
`164
`164
`
`1.
`2.
`
`3.
`4.
`5.
`
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Introduction .
`.
`.
`.
`.
`.
`.
`Current treatment against opiates and alcohol dependency .
`.
`.
`.
`.
`.
`2.1.
`Agonist therapy: methadone and associated problems .
`2.2.
`Partial agonist therapy: buprenorphine and associated problems .
`2.3.
`Antagonist therapy: naltrexone and its mechanism of action .
`.
`.
`Limitations of oral NTX .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Drug delivery: basic principles .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Sustained-release NTX formulations
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5.1.
`Sub-cutaneous formulations
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5.2.
`Injectable formulations .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`5.3.
`Novel implants and depot injections .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Sustained-release systems for NTX delivery .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`6.
`Sustained-release NTX implants
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`7.
`Sustained-release NTX injections .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`8.
`Safety and tolerability of extended-release formulations
`.
`.
`.
`.
`.
`.
`.
`.
`9.
`10. Micelles and microspheres for sustained-release of NTX .
`.
`.
`.
`.
`.
`.
`.
`11.
`Nanogels
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`12.
`Conclusions
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`Acknowledgments
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`References .
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`.
`
`.
`
`⁎ Corresponding author. Tel.: +230 4651347.
`E-mail address: djhurry@uom.ac.mu (D. Jhurry).
`
`http://dx.doi.org/10.1016/j.jconrel.2014.03.046
`0168-3659/© 2014 Elsevier B.V. All rights reserved.
`
`AMN1081
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`1. Introduction
`
`2.1. Agonist therapy: methadone and associated problems
`
`N. Goonoo et al. / Journal of Controlled Release 183 (2014) 154–166
`
`155
`
`Treatment options for heroin addiction has long been dependent
`on three main alternatives namely detoxification, opioid agonist
`(i.e. methadone) and partial agonists (i.e. buprenorphine) mainte-
`nance treatment, and oral NTX. Detoxification followed by long-
`term residential treatment was found to cause some reduction in
`drug use but suffered from problems such as lack of retention in
`treatment and risk of overdose upon discharge [1]. Opioid maintenance
`treatment (OMT) involves the administration of opioid agonist medica-
`tions such as methadone, buprenorphine and medically dispensed
`heroin under supervision [2]. OMT has been effective in decreasing
`mortality rates, morbidity and drug-related criminal activity. How-
`ever, dropout rates remain quite high during the initial months of
`treatment.
`As regards alcohol abuse, detoxification, non-pharmacological
`(psychosocial) treatment methods and pharmacotherapy have not
`been very effective. Disulfiram (Antabuse®), Naltrexone (Revia®),
`and calcium acetylhomotaurinate (Acamprosate®) are the three
`major oral pharmacotherapies used in the treatment of alcoholism.
`Disulfiram is a deterrent medication and makes its ingestion un-
`pleasant. Acamprosate®, a glutamate antagonist has been found
`promising in the treatment of alcoholics [3,4] but present limita-
`tions. For some patients, combination therapy with NTX or disulfi-
`ram have proved to be effective [5].
`The development of long-acting depot formulations of NTX has
`led to improved results such as increased bioavailability and efficacy
`of treatment and is considered as a solution to the problem of non-
`compliance and extensive first pass metabolism associated with
`oral NTX. This has been summarized in two excellent review papers
`[6,7]. In their review, Lobmaier et al. [6] emphasized on NTX depot
`formulations for opioid and alcohol dependence, discussing the
`mode of administration, the pharmacokinetic properties, safety and
`tolerability of the systems. The authors concluded on the need for
`further research on NTX to effectively block clinically relevant
`doses of heroin. Krupitsky et al. [7] summarized the effectiveness
`and safety of long-acting sustained release injectable and implant-
`able formulations of NTX for heroin dependence. The authors con-
`cluded on improved tolerability and effectiveness of long-acting
`sustained-release NTX systems compared to oral NTX. They also
`mention that studies comparing the injectable formulation with
`oral NTX are required. In both reviews, the delivery systems are lim-
`ited to NTX-loaded polymer-based microspheres.
`This article reviews existing naltrexone delivery systems and their
`limitations and presents benefits of emerging nano-based delivery sys-
`tems. In the first part of the review, we present the mechanism of action
`of NTX and its interest as a substitute for methadone followed by an in-
`depth analysis of commercially available NTX formulations with more
`recent references based on clinical trials through 2011 to 2013. We
`have summarized safety and tolerability aspects of extended-release
`formulations to ease access to information. We also stress on new
`nano-based NTX developments such as block copolymer micelles and
`cross-linked nanogels that attract a lot of interest and opens up new
`perspectives for research.
`
`2. Current treatment against opiates and alcohol dependency
`
`Opiates generally refer to any of the narcotic opioid alkaloids found
`as natural products in the opium poppy plant, Papaversomniferum [8].
`Few examples of opiates include heroin and codeine. Opiate drugs
`act both in the central and peripheral nervous systems and opiate-
`dependent patients show impairment in brain functioning [9,10].
`Agonists and partial agonists such as methadone and buprenorphine
`respectively, and antagonists such as NTX have been used in the
`management of opioid dependence.
`
`Methadone was first developed in Germany in 1937. However, its
`use as a substitute for heroin in the management of heroin depen-
`dence was not until 1964 [11]. Methadone has cross tolerance with
`other opioid compounds such as heroin, morphine and codeine and
`can therefore be used as a chemical replacement for the illicit opioid.
`The treatment of opioid addicts with methadone involves an initial
`methadone maintenance program (MMT). MMT is the most widely
`used opioid substitution program for the management of heroin
`dependence and its clinical efficacy has been repeatedly shown by
`several studies [12]. Being long acting, methadone should be admin-
`istered only once daily as opposed to heroin which requires twice or
`thrice daily dose administration. Its oral route of administration sub-
`stantially reduces the potential risks of spreading Hepatitis C or HIV.
`However, methadone therapy has few limitations.
`Methadone therapy is associated with a number of problems. Due to
`its full μ opiate receptor agonist action, there is no limit to the level of re-
`spiratory depression or sedation that methadone can induce. As a result,
`methadone overdose can be lethal, with risk being particularly high
`during the induction period [13]. The combination of methadone with
`other opioid drugs, benzodiazepines or alcohol increases the risks of
`sudden cardiac death [14] and death by anoxic brain injury with pulmo-
`nary edema secondary to respiratory depression [15]. Methadone may
`increase the likelihood of QT interval prolongation [16] and may be as-
`sociated with torsades de pointes [17] that can be fatal.
`As methadone has a long half-life, coming off methadone is asso-
`ciated with a longer period of opioid withdrawal symptoms than
`when coming off heroin. This results in a significant degree of dis-
`comfort in patients who attempt to stop methadone. Methadone is a
`corrective but not a curative treatment for opioid addiction. Newer
`treatments with opioid antagonists like long acting NTX formulations
`need to be explored further as the initial results look promising.
`
`2.2. Partial agonist therapy: buprenorphine and associated problems
`
`Buprenorphine is a partial μ agonist and κ opiate receptor antagonist.
`It is also used in the treatment of opioid dependence and has several po-
`tential benefits over MMT. It is less sedating than methadone due to the
`fact that it is a partial μ receptor agonist. Also, it is associated with lower
`overdose risk since it rarely causes respiratory depression when used
`alone [18]. One way of reducing the abuse liability of buprenorphine
`[19] without affecting its bioavailability has been via the addition of nal-
`oxone hydrochloride to buprenorphine in a ratio of 1:4 (Suboxone,
`Reckitt Benckiser) [20]. Suboxone® was approved in April 2006 by the
`Therapeutics Goods Administration (TGA), and is now largely replacing
`buprenorphine hydrochloride (Subutex®) as the principal formulation
`for ambulatory clinical treatment of opioid dependence. Buprenorphine
`is available in different forms as summarized in Table 1.
`New dosage forms of buprenorphine include transdermal patches
`[22], orodispersible or mucoadhesive buccal films [23]. The transdermal
`buprenorphine patch, Transtec®, first launched in 2001 uses a matrix
`technology whereby buprenorphine is homogeneously incorporated
`in a solid polymer matrix patch [22]. Transdermal buprenorphine
`patches are available in three different dosages with total loading
`doses of 20 mg, 30 mg, and 40 mg which release the drug at a controlled
`rate of 35 μg/h, 52.5 μg/h and 70 μg/h respectively [22]. BUNAVAIL™ is
`the first and only buccal formulation of buprenorphine and naloxone
`[24]. A New Drug Application (NDA) was submitted to the Food and
`Drug Administration (FDA) in 2013 and is currently under review.
`A consensus on the relative superiority of buprenorphine over MMT
`remains elusive. Many studies reveal no significant differences between
`the treatments [25]. Others report significantly higher rates of retention
`in treatment, and abstinence from, or reduction in illicit opiate con-
`sumption among buprenorphine patients than among MMT patients
`[26]. A few studies described more favorable outcomes for MMT than
`
`AMN1081
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`156
`
`N. Goonoo et al. / Journal of Controlled Release 183 (2014) 154–166
`
`Table 1
`Different forms of buprenorphine.
`
`Trade name
`
`Dosage form
`
`Subutex® (buprenorphine)
`Suboxone® (buprenorphine/naloxone)
`Zubsolv® (buprenorphine/naloxone)
`Transtec®
`Butrans®
`Norspan ®
`
`Sublingual tablet (2 mg and 8 mg)
`Sublingual film (4 mg buprenorphine/1 mg naloxone and 12 mg buprenorphine/2.5 mg naloxone)
`Sublingual tablet (2 mg buprenorphine/0.5 mg naloxone and 8 mg buprenorphine/2 mg naloxone)
`Transdermal
`Transdermal (delivering 5, 10 or 20 g/h)
`Transdermal (delivering 5, 10 or 20 g/h)
`
`References
`
`[21]
`
`[22]
`[23]
`
`for buprenorphine in terms of retention, abstinence for at least three
`weeks, opioid-free urine [27], and cost-effectiveness [28]. Nevertheless,
`overall pharmacokinetic features suggest that buprenorphine is safer
`than MMT, with respect to its reduced risk of respiratory depression,
`withdrawal symptoms, and accidental opioid overdose deaths [29]
`and reduced potential for abuse [30].
`
`2.3. Antagonist therapy: naltrexone and its mechanism of action
`
`Narcotic antagonists such as NTX, have been found useful in the
`treatment of both opiate addiction and alcohol dependency [31,32].
`NTX has a chemical structure similar to opiates and can occupy the
`body's opiate receptors in preference to opiates. The ability of NTX
`to effectively antagonize heroin use is unequivocal [33,34]. Studies
`have reported serum NTX levels of 2.8 ng/ml as being effective in
`blocking 500 mg of snorted pure pharmaceutical diamorphine [35],
`serum naltrexone levels N2 ng/ml [34–38] as being effective in
`blocking the effects of 25 mg intravenously administered heroin,
`and others have reported plasma levels of less than 1 ng/ml as
`being capable of antagonizing the effects of 15 mg morphine [39].
`NTX is an opioid receptor antagonist that blocks the reinforcing
`effects of opioids and reduces alcohol consumption and craving.
`Historically, N-allylnorcodeine was the first opioid antagonist-like
`molecule developed in 1915. It acted by blocking the respiratory-
`depressant effects of morphine and heroin. In the 1940s, nalorphine
`was the first reported opioid antagonist but was found to cause dys-
`phoria, discouraging its use in the treatment of opioid intoxication
`and overdose. Naloxone was then developed in 1960 as a less toxic
`antagonist. It did not cause any dysphoria but suffered from short
`duration of action and poor oral bioavailability. To circumvent
`these disadvantages, NTX was developed in 1963 by Endo Laborato-
`ries, which was later acquired by DuPont. It is generally synthesized
`from thebaine (an opiate alkaloid) [40] and was found to have better
`oral bioavailability, a longer duration of action and twice as potent as
`naloxone. Naltrexone hydrochloride is freely soluble in water, slightly
`soluble in ethanol (approximately 96%), and practically insoluble in
`methylene chloride [41]. It is a BCS Class IV drug i.e. it has low solubility
`and low permeability.
`Table 2 gives a summary of the pharmacokinetic data of NTX. NTX
`is FDA-approved for the treatment of alcoholism or opioid addiction
`in the form of commercially available oral tablets e.g. Trexan®,
`Revia®, Depade® or the long-acting, high-dose depot form Vivitrol®
`for intramuscular injection.
`
`Table 2
`Pharmacokinetic data of NTX [37,42].
`
`Chemical formula
`Oral bioavailability
`Metabolism
`Peak concentration
`Half-life
`Duration of action
`Elimination
`Peak plasma level
`
`Naltrexone
`
`C20H23NO4
`Up to 40%
`Hepatic
`1–2 h
`Up to 14 h (oral)
`Up to 24+ h
`Hepatic metabolism and renal excretion
`1 to 2 ng/ml
`
`Studies have revealed that the mesolimbic dopamine system is
`the prime target of addictive drugs. This system originates in the ven-
`tral tegmental area (VTA) of the brain (Scheme 1). Most projection neu-
`rons of the VTA are dopamine-producing neurons. GABA interneurons
`suppress dopamine cell firing resulting in reduced dopamine release.
`Opioids block the inhibitory control exerted by these neurons over the
`VTA dopamine cell bodies resulting in increased VTA dopamine activity,
`thus enhancing brain-reward (reinforcement circuit in the human
`brain) and inducing drug-taking behavior and possibly drug-craving.
`Each addictive drug has a specific molecular target which engages a dis-
`tinct cellular mechanism. The main molecular receptors of opioids are
`μ-OR Gio protein-coupled receptors.
`NTX acts by blocking the μ-opiate receptors, thus reducing crav-
`ing. The precise mechanism for craving reduction has not been de-
`termined yet, but it is likely that NTX causes antagonism of opioid
`pathways to the nucleus accumbens, thereby reducing the total
`amount of dopamine released (Scheme 1). In addition, opioid antag-
`onists like NTX influences other biological systems such as G-
`receptor second messenger systems [43], immune system [44] and
`the HPA axis [45]. NTX is metabolized in the liver into a variety of
`metabolites, with 6-β-naltrexol being the metabolite useful in
`treating drug abuse (Scheme 2). 6-β-naltrexol is believed to act as
`a competitive antagonist at opioid receptors. Cytochrome P450 en-
`zymes, which are involved in the metabolism of methadone or
`buprenorphine do not play a role in NTX metabolism. NTX is largely
`metabolized by the aldo-ketoreductase family of enzymes (AKR1C1,
`1C2 and 1C4) [46] with AKR1C4 being the most efficient [47]. It is be-
`lieved that a polymorphism of the AKR1C4 enzyme is responsible for
`inter-individual variability in 6-β-naltrexol levels and could be used
`to explain the efficacy of and compliance with NTX treatment [46].
`Due to its higher potency compared to naloxone and cyclazocine,
`NTX is considered as the most promising narcotic antagonist used for
`the treatment of narcotic addiction [48,49]. A minimum plasma level
`of NTX of 1 ng/ml is required for blocking clinically relevant doses (e.g.
`25 mg) of intravenously administered heroin [50]. Evaluation of a pro-
`gram where cognitive behavioral therapy (CBT) and/or NTX were
`used over 12 weeks showed that addition of NTX significantly improved
`the abstinence rate (36.1% CBT against 62.6% CBT + NTX) [51].
`However, oral NTX (Revia® tablets) has been associated with
`high early dropout rates. It was shown that 37% of patients discontin-
`ue daily oral NTX by 12 weeks [52] and more than 80% discontinue
`use by 6 months [53]. As demonstrated by several studies, compli-
`ance is critical for the efficacy of NTX [54]. Moreover, orally adminis-
`tered NTX has poor bioavailability due to high hepatic metabolism
`(98%) and a wide fluctuation in drug plasma concentration occurs
`with orally administered NTX [55]. Indeed, a review of the effective-
`ness of oral naltrexone maintenance for the treatment of opioid de-
`pendence concluded that there was insufficient evidence to justify
`the use of NTX in maintenance treatment of opioid addicts [56].
`
`3. Limitations of oral NTX
`
`As mentioned earlier, NTX is available commercially as tablets
`for oral administration. However, they have several pharmaco-
`therapeutic limitations. First of all, more than 98% of the drug is me-
`tabolized in the liver and very small amount reaches the brain. Due
`
`AMN1081
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`N. Goonoo et al. / Journal of Controlled Release 183 (2014) 154–166
`
`157
`
`VTA of brain consisting of
`GABAergic and dopaminergic
`neurons
`
`Alcohol stimulates the
`release of β-
`endorphins
`
`which bind to μ-
`opioid receptors
`
`thus
`releasing
`dopamine
`
`…
`
`…
`
`A
`
`B
`
`Alcohol stimulates the
`release of β-
`endorphins
`
`thereby
`preventing
`dopamine
`release
`
`…
`
`6- β-naltrexol blocks the
`binding of β-endorphins to
`μ-opioid receptors
`C
`
`β-endorphins
`
`6- β-naltrexol
`
`μ-opioid receptor
`
`dopamine
`
`Scheme 1. Schematic representation of interplay between GABAergic and dopaminergic
`neurons in (A) absence of drug of abuse and its antagonist, (B) presence of drug of
`abuse only and (C) presence of both drug of abuse and its antagonist.
`
`to extensive first pass metabolism, the concentration of naltrexone and
`the active metabolite, 6-β-naltrexol peaks within the first hour after
`oral dosing, followed by a steady decline each day during treatment
`[57]. This explains the need for the development of a system whereby
`NTX bypasses the liver i.e. an injectable long-acting drug delivery
`system. Such a system will enable the maintenance of a constant and
`predictable drug plasma concentration. According to a study conducted
`
`HO
`
`O
`
`O
`
`OH
`
`N
`
`HO
`
`O
`
`HO
`
`OH
`
`N
`
`Naltrexone (NTX)
`
`6-β-naltrexol
`
`Scheme 2. Metabolism of NTX to 6-β-naltrexol.
`
`by Verebey et al. [55] among alcoholics, drug plasma levels fluctuates
`much with orally administered NTX. In fact, a 100 mg naltrexone dose
`provided 96%, 86.5% and 46.6% blockade at 24 h, 48 h and 72 h respec-
`tively. Moreover, the use of oral NTX places the onus on the patients as
`to whether to take the medications or not and very often, they do not
`comply with the required frequency. Studies have also shown that a
`comparatively low proportion of patients choose to start NTX treatment
`[58]. Among those who do, many drop out early; one quarter after a few
`days [33] and as many as half in the first few weeks of treatment [59].
`This is a major problem given that several studies have demonstrated
`that missing even a few doses of NTX could lead to full relapse into opi-
`oid use and discontinuation of the treatment, despite intensive clinical
`interventions [54,60].
`
`4. Drug delivery: basic principles
`
`Drug delivery systems (DDS) may be differentiated according to the
`way the drug is administered or released. They may be administered
`through oral or parenteral (intravenous, intramuscular, subcutaneous,
`intradermal or intraperitoneal) routes [61].
`DDS can broadly be classified as immediate release and modified re-
`lease dosage forms. Modified-release systems can be further divided
`into delayed-, extended- and targeted-release systems. Furthermore,
`extended-release systems can be divided into sustained- and controlled
`release systems [61] (Fig. 1).
`Sustained release systems maintain the rate of drug release over a
`sustained period of time [61]. Sustained release systems may be either
`in the form of reservoir or matrix systems. Reservoir systems often
`follow a zero-order kinetics (linear release as a function of time) while
`matrix systems often follow a linear release as a function of the square
`root of time. Sustained release systems offer several advantages such
`as reduced fluctuations in drug concentrations, and reduced total
`dose. Also, the patient does not require taking the drug frequently and
`therefore resolves the issue of non-compliance.
`Controlled-release systems are different from sustained-release
`ones [61]. They are designed to maintain specific plasma concentrations,
`independent of the biological environment of the application site [61,
`62]. Another major difference is that sustained-release forms are often
`restricted to oral dosage forms. On the other hand, controlled-release
`systems are used in a variety of administration routes, including trans-
`dermal, oral and vaginal administration [61].
`Release from oral NTX tablets may be termed as a burst release,
`resulting in fluctuating plasma concentrations during the day (Fig. 2).
`NTX concentration peaks within the first hour of oral dosing followed
`by a fairly rapid decline in plasma levels to below the minimum thera-
`peutic levels (2 ng/ml) within 8 h of dosing [63]. The use of a sustained
`release NTX formulation will result in slow NTX release, avoiding the
`peaks and troughs associated with daily drug administration, while
`maintaining continuous therapeutic plasma levels for an extended
`time frame. This “smoothing out” of drug levels in the blood may de-
`crease the possibility of occurrence of adverse events associated with
`peaks, and improve efficacy by avoiding drug concentration troughs.
`
`AMN1081
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`N. Goonoo et al. / Journal of Controlled Release 183 (2014) 154–166
`
`Minimum toxic concentration
`
`Table 3
`Kinetic models used for analysis of drug release data.
`
`Model Name
`
`Model
`
`Overall mean percent error
`
`158
`
`concentration
`
`Drug
`
`Zero order
`First order
`Higuchi
`Hixson–Crowwell
`Power law
`
`Mt = M0 + k0t
`log Ct = log C0 − Kt/2.303
`Mt = KHt1/2
`M01/3 − Mt
`
`1/3 = κ t
`ln F = ln Kp + p ln t
`
`18.28
`16.41
`10.65
`26.63
`7.66
`
`Mt: amount of drug dissolved in time t.
`M0: initial amount of drug in the solution.
`k0: zero-order release constant.
`Ct: concentration of drug dissolved in time t.
`C0: initial concentration of drug.
`K: first order rate constant.
`t: time.
`KH: Higuchi dissolution constant.
`κ: constant incorporating the surface-volume relation.
`F: fraction of drug released at time t.
`p, Kp : parameters of the model.
`
`are given in Table 4. The t50% value obtained for the more hydrophobic
`PEO–MMA copolymer (1:4) suggests a more sustained release com-
`pared to the PEO–MMA copolymer (1:1).
`
`ð6Þ
`
`−1 ¼ m
`tb
`
`1 F
`
`The use of kinetic models helps to elucidate release mechanisms, which
`can in turn be useful to control drug release. The mathematical models
`discussed above can help optimize existing systems and ultimately de-
`sign a polymer-based therapeutic system with the drug released at the
`required rate and concentration.
`
`5. Sustained-release NTX formulations
`
`An alternative NTX maintenance delivery against the problem of
`non-compliance involves injection or surgical insertion of a sustained
`release preparation of NTX, avoiding the gastro-intestinal route. This
`removes the need for daily oral NTX.
`
`5.1. Sub-cutaneous formulations
`
`The concept of sustained release preparations of NTX is not new.
`Beginning in the mid-1970s, a number of depot formulations of NTX
`were developed. While showing promising NTX release patterns,
`and being of ‘likely biodegradable materials’, most had unacceptable
`tissue compatibility. For example, Chiang et al. [67] conducted one of
`the early studies of sustained release NTX in normal, healthy volun-
`teers implanted subcutaneously with naltrexone-copolymer (90% L-
`lactic acid and 10% glycolic acid) beads. Following an initial burst of
`release, this formulation yielded relatively constant plasma levels
`of NTX (0.3–0.5 ng/ml) for up to 1 month. Data indicated that this
`NTX preparation had unacceptable levels of biocompatibility, with
`two of the three human subjects implanted with the naltrexone-
`copolymer (90% L-lactic acid and 10% glycolic acid) beads having
`
`Table 4
`Summary of parameters obtained for NTX release using the reciprocal powered method
`[66].
`
`Nanosystem
`
`PEO–MMA copolymer (1:1)
`PEO–MMA copolymer (1:4)
`
`Na
`
`6
`17
`
`R2
`
`0.895
`0.650
`
`E
`
`3.0
`10.5
`
`m
`
`1.967
`3.559
`
`b
`
`0.603
`0.431
`
`t50% (h)
`
`3.1
`19.0
`
`Controlled release
`Delayed release
`
`Immediate release
`Burst release
`
`Sustained release
`
`Minimum effective concentration
`
`Time/ hours
`
`Fig. 1. Drug release kinetics.
`
`Drug release may be modeled using different models as shown in
`Table 3 [64]. The R2 values are used to check which model best fits the
`release system.
`Polymer based drug delivery systems may be categorized as
`diffusion-controlled, solvent-activated (swelling or osmotically con-
`trolled), chemically controlled or externally-triggered (e.g. pH, tem-
`perature) [65].
`Immediate-release, modified-release, extended-release and delayed-
`release have been defined by the FDA. However, no definitions have
`been provided for targeted or controlled release [61].
`Barzegar-Jalali et al. reported on a general model applicable to
`multi-mechanistic release from nanoparticles (Eq. (6)) [66]. Parameters
`obtained from this model may be used to compare different delivery
`systems of a given drug as well as correlating with bioavailability data.
`Indeed, the release half life, t50% can be used to compare release rates
`of different systems. The values of the different parameters obtained
`for NTX-loaded hydrolyzable crosslinked nanoparticles (using Eq. (6))
`
`Fig. 2. (A) Typical profile of plasma NTX levels over 24 h following a 50 mg oral dose in
`humans. (B) Simulation of the daily fluctuations in plasma levels of NTX over the course
`of a month, assuming the patient adheres to the daily dosing of oral NTX.
`Reprinted with permission from [63].
`
`N: number of data in each set.
`E: percent error.
`F: fraction of drug released in time t.
`m, b: parameters of the model.
`
`AMN1081
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`

`

`N. Goonoo et al. / Journal of Controlled Release 183 (2014) 154–166
`
`159
`
`them removed at approximately 3 to 4 weeks due to marked inflam-
`matory reactions or other local tissue irritation. Chiang concluded
`that this result “may preclude the clinical use of this particular prep-
`aration of beads” [67].
`
`noncompliance issue associated with oral NTX, and produce good
`treatment outcomes, the requirement that patients return for re-
`treatment every 30 days [74,85] is associated with high attrition
`rates post 60 days (25–36%) and limits clinical efficacy.
`
`5.2. Injectable formulations
`
`6. Sustained-release systems for NTX delivery
`
`Newer formulations of sustained-release NTX have provided
`more promising results. Injectable formulations of NTX, such as
`those produced by Biotek, Inc. (Depotrex®; [68]), Drug Abuse Sci-
`ences (Naltrel®; [69]) and Alkermes, Inc. (Vivitrex®; [70]) appear
`to produce both clinically relevant plasma concentrations of NTX
`(1–2 ng/ml) for approximately 3–6 weeks, with clinically acceptable
`incident level of tissue reactivity. For example, an injectable depot
`formulation of NTX (Depotrex®, 192 mg, 384 mg NTX base) antago-
`nized the effects of intravenously-administered heroin (0–25 mg)
`for 3–5 weeks, depending on NTX dose. This study demonstrated
`that Depotrex® was safe, effective, and well tolerated in opioid
`abusers who were not seeking treatment for their drug use. A subse-
`quent “proof-of-concept” clinical trial of Depotrex® in treatment-
`seeking heroin abusers showed a robust, dose-related increase in
`treatment retention, supporting the effectiveness of NTX in antago-
`nizing the objective and subjective effects of heroin [71].
`
`5.3. Novel implants and depot injections
`
`Recently, extended-release formulations that release NTX for
`1–7 months have become available for clinical use. Such systems
`consist of compressed NTX or NTX/polymer/copolymer administered
`sub-cutaneously or intra-muscularly [72]. Most studies have indicated
`the effectiveness of these systems which have acceptable adverse
`event profiles [50]. A 30 day injectable NTX, Vivitrol® was approved
`by the FDA for treatment of alcohol dependence or opioid dependence
`in 2006 and 2010, respectively. It was shown that patients who received
`400 mg treatment during 28 days had a blood NTX level of 1.23 ng/ml
`[73]. A larger 6-month trial of Vivitrol®: (205 received 380 mg injec-
`tions, 210 received 190 mg injections, 209 received placebo injec-
`tions, plus psychosocial intervention) also reported a reduction of
`25% (p = 0.03) in heavy drinking days in the 380 mg NTX, and 17%
`(p = 0.07) in the 190 mg dose compared to placebo recipients. How-
`ever 36% of patients failed to complete the 6-month course of
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket