Content » Vol 92, Issue 5

Review

Systemic Kappa Opioid Receptor Agonists in the Treatment of Chronic Pruritus: A Literature Review

Ngoc Quan Phan1, Tobias Lotts1, Attila Antal2, Jeffrey D. Bernhard3 and Sonja Ständer1

1Competence Center Chronic Pruritus, Department of Dermatology, University Hospital Münster, Münster, 2Department of Dermatology and Allergology Ludwig-Maximilians-University, München, Germany, and 3Department of Dermatology, University of Massachusetts Medical School, Worcester, Massachusetts, USA

Chronic pruritus is frequently refractory to currently available treatments. Studies suggest that pruritus may arise from an imbalance of the mu- and kappa-opioid receptor system activity in either the skin or the central nervous system. Stimulation of kappa-opioid receptors by their agonists inhibits pruritus in both animals and humans. The antipruritic effect of kappa-opioid receptors agonists can currently be assumed to be related to their binding to kappa-opioid receptors on keratinocytes and cutaneous and/or central itch neurones. To date, several case reports and 2 controlled trials have demonstrated a beneficial effect of systemic kappa-opioid receptor agonists in the treatment of uraemic pruritus, prurigo nodularis, paraneoplastic and cholestatic pruritus. Nalfurafine hydrochloride (Remitch®), a selective kappa-opioid receptor agonist, is approved for the treatment of chronic pruritus in Japan. The aim of this review is to provide an overview of the promising role of kappa- opioid receptors and their agonist in the pathophysiology and treatment of pruritus. Key words: itch; therapy; kappa opioid receptor antagonist; mu-opioid receptor.

(Accepted December 30, 2011.)

Acta Derm Venereol 2012; 92: XX–XX.

Sonja Ständer, Competence Center Chronic Pruritus, Department of Dermatology, University Hospital Münster, Von-Esmarch-Strasse 58, DE-48149 Münster, Germany. E-mail: sonja. staender@ukmuenster. de

Chronic pruritus is frequently refractory to treatment. Accordingly, a symptomatic antipruritic therapy that would inhibit the induction, transmission, or perception of itch is badly needed. During the past 10 years, opioid receptors have been identified as a target in the treatment of pruritus. Mu- and kappa-opioid receptors are expressed in the skin and central nervous system (CNS) (1). Mu-opioid receptors (MOR) antagonists are effective in chronic pruritus of various origins (1). MOR and kappa-opioid receptors (KOR) mediate opposite effects (2). For example, activation of MOR inhibits pain, while activation of KOR inhibits itch. The newly developed KOR agonists have demonstrated in experimental and clinical studies antipruritic effects in antihistamine-resistant and antihistamine-sensitive pruritus in animals and humans (3, 4). Given that an imbalance of the endogenous opioid system has been considered as a pathomechanism in uraemic pruritus, antipruritic effects of KOR agonists have been investigated in randomized controlled trials (RCT) in this indication (5–8). The major advantage of the KOR agonists over MOR antagonists seems to be the fewer rates of up-to-date reported adverse events. MOR antagonists show a high rate of troublesome side-effects (1), hampering their application mainly in elderly patients. The aim of this paper is to provide an overview of the current knowledge of KOR and their role in pruritus as well as the evidence of antipruritic potency of KOR agonists and their possible use in the treatment of chronic pruritus.

METHODS

A systemic search for reports of KOR and KOR agonist in experimental animal models and studies in humans was carried out. The major electronic databases (PubMed, Ovid and DIMDI) were searched using the free-text key words kappa opioid receptor or kappa opioid receptor agonist, butorphanol, nalfurafine and/or skin, pruritus, itch, and several combinations of these words. Searching was carried out without time and language restriction, and without contacting manufacturers or authors. In addition, information on these two drugs from current data sheets was included.

RESULTS

A total of 3,183 papers (until August 2011) with the key word “kappa opioid receptor agonist” were found, mostly addressing the analgesic properties of KOR agonists. A combination of the key words “”kappa opioid receptor agonist” and “itch” or “pruritus” resulted in 28 papers, of which only 7 were case reports, case studies or randomized trials on antipruritic effects of KOR agonists in humans. In addition, we included papers on mechanisms of action, e.g. in animal models and experimental models as well as information from current data sheets of KOR agonists, finally resulting in 74 references.

Kappa opioid receptor expression

Opioid receptors (ORs) are a member of the pertussis toxin-sensitive heterotrimeric Gi/Go protein-coupled receptor family with a 7 transmembrane domain and belong to the rhodopsin subfamily (9, 10). Three major classes of opioid receptors have been identified: KOR, MOR and delta-opioid receptors (DOR) (11–13). MOR is specific for the ligand beta-endorphin, DOR for enkephalins and KOR for dynorphins (Dyn) (14), but no ligand is completely specific for any receptor (15). It is assumed that 3 different subtypes of KOR (k1, k2 and k3) exist, based on ligand receptor binding studies, but only one has been identified so far (GenBank Acc. No. U11053) (16–22).

KOR is expressed in the CNS as well as in the periphery. In the CNS, KOR are localized in axons in the medial prefrontal cortex (23), in the nucleus accumbens, hypothalamic nuclei, periaqueductal gray (PAG), nucleus of the solitary tract, ventral tegmental area, amygdala, neocortex and supraspinal nucleus (24–26) and in the dorsal horn of the spinal cord (27). Interestingly, in the dorsal horn of the spinal cord, KOR may be linked with the cannabinoid system (28). Systemic application of the cannabinoid receptor agonist delta-9-tetrahydorcannabinol (THC) was demonstrated to inhibit nociceptive reflexes, possibly resulting from an inhibition of dorsal horn neurones through a KOR-dependent mechanism (28). KOR-expressing neurones are also located in the dorsal and trigeminal root ganglia (29), which is an important link to the peripheral nervous system. In the periphery, KOR has been detected in nerve terminals of muscles, joints and viscera (30). Expressed widely in the CNS and peripheral nervous system, the KOR system was shown to play an important role in anti-nociception (31), dysphoria (32, 33) and water diuresis (31, 33, 34).

KOR are expressed in human epidermal keratinocytes (35, 36) as well as dermal fibroblasts (36, 37) and mononuclear cells (37) as demonstrated in cell cultures (e.g. HaCaT, HNK) and skin biopsies on mRNA and protein levels. KOR expression in subepidermal nerve fibres was demonstrated by immunohistochemical staining (36). The corresponding KOR ligands DynA (1–17) and DynA (1–8) were identified in the epidermis (35). Studies suggest that KOR contribute to proliferation and differentiation of keratinocytes (38). For example, KOR knockout mice revealed epidermal hypotrophy and slightly enhanced cutaneous nerve fibre density after induction of dry skin dermatitis (38). The epidermis was significantly thinner than in wild-type mice (38). In addition, real-time polymerase chain reaction (PCR) showed increased KOR expression in epidermal keratinocytes and fibroblasts in hypertrophic scars in comparison with normal skin (36), which may contribute to the increased nociception in scars. Interestingly, a recent paper demonstrated that KOR expression was significantly decreased in the epidermis of itchy psoriatic patients compared with healthy controls (39). Psoriasis shows an epidermal hyperplasia, but no thinned epidermis. The reduced KOR expression argues against a role of KOR in differentiation of keratinocytes in patients with psoriasis. Further studies need to address the role of KOR in human keratinocyte biology (39). KOR are expressed on blood derived dendritic cells (40). In cell cultures (KOR transfected Jurcat T cells), activation of KOR down-regulates cytokines and chemokines in inflammatory cells, and thus could induce an anti-inflammatory response (41). This is of great interest for inflammatory diseases presenting with pruritus, such as atopic dermatitis (AD).

Role of kappa opioid receptor in pruritus

Like MOR, KOR plays a role in the sensation of itch in animals and humans (34, 42). Systemic administration of the selective and potent KOR antagonist norbinaltorphimine induced scratching in mice (42). In contrast, activation of itch neurones in the superficial layer of the dorsal horn of the spinal cord was shown to be inhibited by the KOR agonist TRK-820 (nalfurafine) in mice (43). Consequently, oral (3, 44) or subcutaneous (45, 46) administration of nalfurafine resulted in significant reduction of substance P (SP) or histamine-induced pruritus in wild-type mice. In addition, spontaneously developing scratching behaviour in NC/Nga mice could be suppressed by nalfurafine (3, 44–46). Scratching behaviour was reduced in KOR knockout mice after dry skin induction in comparison with wild-type mice (38). In a model of autoimmune-induced pruritus in aged MRL/lpr mice, oral nalfurafine also inhibited scratching behaviour (47). In a rat model of cholestatic pruritus, nalfurafine reduced scratching behaviour significantly (48). Intravenous application of TRK-820 suppressed scratching induced by systemic application of morphine in rhesus monkeys (49).

Together, KOR are involved in the pathophysiology of pruritus not only by their expression in the CNS, but also by their presence in the skin. For example, application of the peripherally acting KOR agonist ICI 204,448 inhibited chloroquine-induced pruritus in mice suggesting a peripheral pathway in itch suppression (50). Moreover, it was demonstrated that KOR immunostaining is down-regulated in the epidermis of atopic dermatitis (AD) and itchy psoriasis patients (35, 39). Psoralen plus ultraviolet A (PUVA) therapy did not lead to normal KOR levels in AD, but an increase in the KOR ligand DynA (35). Interestingly, KOR is up-regulated in the skin of patients with painful diseases such as fibromyalgia and painful scars (36, 51).

Kappa opioid receptor agonists in clinical use

The antipruritic effects of several artificial KOR agonists have been studied in rodents and monkeys, including models of histamine-, SP- and morphine-induced itch (43–49). Besides nalfurafine, all KOR agonists currently approved for therapeutic use in humans are mixed KOR agonists/MOR antagonists with similar receptor affinity-dependent side-effects of varying intensity. Nalbuphine, butorphanol and pentazocine are well known for their beneficial effects in acute morphine-induced pruritus (52–54). Only butorphanol and nalfurafine have been applied in chronic pruritus (Table I).

Table I. Effect of kappa-opioid receptors (KOR) agonists in chronic pruritus

Diagnosis

KOR agonists in studies

(dosage; ref.)

Study design

n

Compared with

Effect

Uraemic pruritus

Nalfurafine (5 μg i.v. 3/week for 2 or 4 weeks; 4)

Nalfurafine (2.5 and 5 μg p.o. for 14 days; 8)

RCT

RCT

144

337

Placebo

Placebo

Significant improvement in pruritus (p < 0.05)

Significant decrease in pruritus intensity on VAS (p < 0.0001)

Prurigo nodularis

Butorphanol (1 mg/day; 70)

CR

1

Improvement

Primary biliary cirrhosis

Butorphanol (1 mg/day; 70)

CR

1

Improvement

Idiopathic pruritus in elderly patients

Butorphanol (1 mg/day; 70)

CR

1

Improvement

Non-Hodgkin’s lymphoma

Butorphanol (1 mg/day; 70)

CR

1

Improvement

Pruritus in chronic renal failure, diabetes (perforating collagenosis)

Butorphanol (1 mg/day; 70)

CR

1

Improvement

CR: case report; RCT: randomized controlled trial; VAS: visual analogue scale; i.v.: intravenous; p.o.: per oral.

Nalfurafine

Nalfurafine (TRK-820) was developed in 1998 (55) and is a full agonist for the KOR, a partial agonist for the MOR and low-affinity antagonist for the nociceptin receptor (56). Compared with other synthetic KOR agonists, nalfurafine showed higher selectivity for KOR (46, 56). Nalfurafine has anti-nociceptive effects, as demonstrated in mice (e.g. hot plate test, tail flick test, tail pressure test) (57) and monkeys (58). The antipruritic effects of nalfurafine were investigated for the first time in 2002 (3). Pruritus induced by SP- and histamine was significantly inhibited by orally administered nalfurafine in mice, whereas antihistamines did not inhibit SP-induced itching (3). In the first multicentre, randomized, double-blind placebo-controlled trial, Wikström et al. (4) demonstrated significant clinical benefit of nalfurafine in humans with uraemic pruritus (n = 144) (Table I). The finding of reduced pruritus intensity was confirmed later in a similar cohort of patients, i.e. patients with uraemic pruritus, (n = 337) in a randomized placebo-controlled trial (8). Patients responded quickly to nalfurafine, after the first 7 days of treatment a considerable change in VAS was observed. Interestingly, the change in VAS was larger in treatment days 8–14 than in the first 7 days. This observation was independent of the dosage. In fact, 2.5 µg seemed to be almost equally effective as 5 µg. Uraemic itch is defined as a localized or generalized, chronic pruritus of almost unknown pathophysiology in patients with chronic renal failure and/or undergoing haemodialysis (4, 5). Given that an elevation of endogenous opioids has been postulated as a contributory factor, the antipruritic effects of nalfurafine in this indication may be specifically targeting the pathophysiology (4, 59–61). Nalfurafine is licensed only in Japan for the therapy of uraemic pruritus. There are studies on the use of nalfurafine in other pruritic diseases in animal models (e.g. atopic dermatitis) (44), but none on humans are available to date.

Evaluation of adverse events demonstrated nalfurafine to be well tolerated in clinical use. Common adverse drug reactions are mediated by the CNS (insomnia, somnolence, vertigo and headache) and the gastrointestinal system (constipation, nausea and vomiting). All events were transient and resolved without therapy discontinuation (4, 8) (Table II). There were no remarkable changes in haematology and blood chemistry or in vital signs/electrocardiography (8). Moderate transient increased prolactin, decreased blood thyroid stimulating hormone (TSH), decreased free testosterone, elevated eosinophils, transient elevation of aspartate aminotransferase and alanine transaminase were some of the adverse events observed (4, 8). Neither addiction nor symptoms of withdrawal were noted in nalfurafine-treated patients. Being a partial agonist for MOR and DOR, nalfurafine significantly reduced physical dependence on morphine in mice and humans (62).

Table II. Side-effects of therapy with the selective kappa-opioid receptor agonist nalfurafine

Adverse drug reactions (ADR) in the phase III study, as occurred in 337 patients (8)

Incidence of > 3%

Insomnia (14%)

Constipation (7%)

Somnolence (4.5%)

Moderate ADR in individual cases

Anorexia

Headache

Pruritus

Sudden hearing loss

Hypertension

Vomiting

Nausea

Transient mood alterations (elevated mood, feeling abnormal)

Laboratory findings

Decreased TSH

Increased eosinophils

Transient increase in prolactin

Decrease in free testosterone

TSH: thyroid stimulating hormone.

Butorphanol

Butorphanol, developed in the 1970s, is an injectable or intranasally applicable mixed KOR agonist/MOR antagonist, with analgesic potency higher than morphine, pethidine or pentazocine, and is administered to patients with acute postoperative pain (63–65). It relieves acute morphine-induced pruritus even in children (66, 67). However, prophylactic application of butorphanol before application of morphine does not prevent induction of pruritus (68). Moreover, butorphanol itself can induce acute pruritus (69) (Table III). An open-label uncontrolled study of butorphanol in single patients reported that the antipruritic effect of this drug sets in 1–4 days after nasal administration in elderly, primary biliary cirrhosis and non-Hodgkin’s lymphoma-associated pruritus (70). An impaired balance between MOR and KOR systems is suggested to have a role in cholestatic pruritus (71); furthermore, intranasal butorphanol was recommended in cholestatic pruritus, but not investigated in trials (72). A formulation containing 2 mg butorphanol tartrate in 100 µl purified water encapsulated into multilamellar phospholipid vesicles was topically used in rats and could be shown to penetrate the skin with low plasma levels (73). This formulation could potentially be used in the future to treat circumscribed pruritus and thus avoid systemic side-effects. Adverse effects of butorphanol are, for example, dizziness, sedation and nausea (64, 74). As with other morphine-like agents, respiratory depression can occur that is reversible in healthy subjects with moderate doses (up to 0.8 mg) of naloxone (64). Psychotomimetic reactions have been reported. Butorphanol is not recommended for use in patients dependent on narcotics. It should be used with caution in patients with increased intracranial pressure, disorders of respiratory function or control. In patients with hepatic or renal impairment, the initial dose of butorphanol should generally be half the recommended adult dose (e.g. 0.5 mg i.v.). Reproduction studies in animals (mice, rats and rabbits) did not reveal any teratogenic potential of butorphanol; however, it should be used with caution in pregnant and nursing women. There have been rare reports of infant respiratory distress/apnoea following the administration of butorphanol injection during labour. Mixed KOR agonists are not recommended for use in patients below 18 years of age.

Table III. Side-effects of therapy with mixed-action Mu-opioid receptor antagonist/kappa-opioid receptor agonist (butorphanol, nalbuphine and pentazocine)

Side-effects

General (body as a whole)

Asthenia/lethargy, dizzinessa, drowsiness, sensation of heata, somnolencea

Digestive/urogenital

Anorexia, constipation, dry mouth, impaired urination nauseaa, stomach pain, vomitinga

Cardiovascular

Hypertension, hypotension, palpitations, syncope, tachycardia, vasodilatation

Respiratory

Bronchitis, cough, dyspnoea, epistaxis, oedema, infection, nasal congestiona (butorphanol), nasal irritation, pharyngitis, rhinitis, sinus congestion, sinusitis, upper respiratory

Neurologic

Abnormal dreams, agitation, anxiety, confusion, dysphoria, euphoria, floating feeling, hallucinations, hostility, headache, insomniaa, nervousness, paraesthesia, tremor, withdrawal symptoms

Special senses

Blurred vision, ear pain, tinnitus, unpleasant taste

Skin/allergic

Flush, hives, pruritus, sweating/clammy

aFrequent appearance.

CONCLUSION

KOR play an important role in pruritus and skin biology. Therefore, application of KOR agonists as systemic, and probably also topical, agents is a promising therapeutic approach to chronic pruritus. Clinical studies have demonstrated the efficacy of the KOR agonist nalfurafine in uraemic pruritus. Evidence from animal models and morphological studies in humans suggests that KOR agonists might also be effective in pruritus of other origin, for example atopic dermatitis.

ACKNOWLEDGEMENT

We thank Rajam Csordas for assistance in the preparation of the manuscript. The work was supported by Dr. August Wolff GmbH&Co.KG Arzneimittel.

REFERENCES

  • Phan NQ, Bernhard JD, Luger TA, Ständer S. Antipruritic treatment with systemic μ-opioid receptor antagonists: a review. J Am Acad Dermatol 2010; 63: 680–688.
  • Pan ZZ. mu-Opposing actions of the kappa-opioid receptor. Trend Pharmacol Sci 1998; 19: 94–98.
  • Togashi Y, Umeuchi H, Okano K, Ando N, Yoshizawa Y, Honda T, et al. Antipruritic activity of the kappa-opioid receptor agonist, TRK-820. Eur J Pharmacol 2002; 435: 259–264.
  • Wikström B, Gellert R, Ladefoged SD, Danda Y, Akai M, Ide K, et al. Kappa-opioid system in uremic pruritus: multicenter, randomized, double-blind, placebo-controlled clinical studies. J Am Soc Nephrol 2005; 16: 3742–3747.
  • Mettang T, Pauli-Magnus C, Alscher DM. Uraemic pruritus – new perspectives and insights from recent trials. Nephrol Dial Transplant 2002; 17: 1558–1563.
  • Ikoma A, Steinhoff M, Ständer S, Yosipovitch G, Schmelz M. The neurobiology of itch. Nat Rev Neurosci 2006; 7: 535–547.
  • Kumagai H, Matsukawa S, Utsumi J, Saruta T. Prospects for a novel kappa-opioid receptor agonist, TRK-820, in uremic pruritus. In: Yosipovitch G, editor. Itch. Basic mechanisms and therapy. New York: Marcel Dekker, 2004: p. 279–286.
  • Kumagai H, Ebata T, Takamori K, Muramatsu T, Nakamoto H, Suzuki H. Effect of a novel kappa-receptor agonist, nalfurafine hydrochloride, on severe itch in 337 haemodialysis patients: a Phase III, randomized, double-blind, placebo-controlled study. Nephrol Dial Transplant 2010; 25: 1251–1257.
  • Tseng RJ, Padgett DA, Dhabhar FS, Engler H, Sheridan JF. Stress-induced modulation of NK activity during influenza viral infection: role of glucocorticoids and opioids. Brain Behav Immun 2005; 19: 153–164.
  • Schwartz TW, Holst B. Molecular structure of G-protein-coupled receptors. In: Foreman JC, Johansen T, editors. Textbook of receptor pharmacology. New York: CRC Press, 2002: p. 81–110.
  • Satoh M, Minami M. Molecular pharmacology of the opioid receptor. Pharmacol Ther 1995; 68: 343–364.
  • Zadina JE, Martin-Schild S, Gerall AA, Kastin AJ, Hackler L, Ge LJ, et al. Endomorphins: novel endogenous mu-opiate receptor agonists in regions of high mu-opiate receptor density. Ann NY Acad Sci 1999; 897: 136–144.
  • Stefano GB, Goumon Y, Casares F, Cadet P, Fricchione GL, Rialas C, et al. Endogenous morphine. Trends Neurosci 2000; 23: 436–442.
  • Goldstein A, Naidu A. Multiple opioid receptors: ligand selectivity profiles and binding site signatures. Mol Pharmacol 1989; 36: 265–272.
  • Kosterlitz HW. The Wellcome Foundation lecture, 1982. Opioid peptides and their receptors. Proc R Soc Lond B Biol Sci 1985; 225: 27–40.
  • Kosterlitz HW, Paterson SJ, Robson LE. Characterization of the kappa-subtype of the opiate receptor in the guinea-pig brain. Br J Pharmacol 1981; 73: 939–949.
  • Zukin RS, Eghbali M, Olive D, Unterwald EM, Tempel A. Characterization and visualization of rat and guinea pig brain kappa opioid receptors: evidence for kappa 1 and kappa 2 opioid receptors. Proc Natl Acad Sci USA 1988; 85: 4061–4065.
  • Clark JA, Liu L, Price M, Hersh B, Edelson M, Pasternak GW. Kappa opiate receptor multiplicity: evidence for two U50, 488-sensitive kappa 1 subtypes and a novel kappa 3 subtype. J Pharmacol Exp Ther 1989; 251: 461–468.
  • Rothman RB, Bykov V, de Costa BR, Jacobson AE, Rice KC, Brady LS. Interaction of endogenous opioid peptides and other drugs with four kappa opioid binding sites in guinea pig brain. Peptides 1990; 11: 311–331.
  • Zhu J, Chen C, Xue JC, Kunapuli S, DeRiel JK, Liu-Chen LY. Cloning of a human kappa opioid receptor from the brain. Life Sci 1995; 56: 201–207.
  • Mansour A, Hoversten MT, Taylor LP, Watson SJ, Akil H. The cloned mu, delta and kappa receptors and their endogenous ligands: evidence for two opioid peptide recognition cores. Brain Res 1995; 700: 89–98.
  • Simonin F, Gavériaux-Ruff C, Befort K, Matthes H, Lannes B, Micheletti G, et al. Kappa-opioid receptor in humans: cDNA and genomic cloning, chromosomal assignment, functional expression, pharmacology, and expression pattern in the central nervous system. Proc Natl Acad Sci USA 1995; 92: 7006–7010.
  • Svingos AL, Colago EE. Kappa-Opioid and NMDA glutamate receptors are differentially targeted within rat medial prefrontal cortex. Brain Res 2002; 946: 262–271.
  • Tempel A, Zukin RS. Neuroanatomical patterns of the mu, delta, and kappa opioid receptors of rat brain as determined by quantitative in vitro autoradiography. Proc Natl Acad Sci USA 1987; 84: 4308–4312.
  • George SR, Zastawny RL, Briones-Urbina R, Cheng R, Nguyen T, Heiber M, et al. Distinct distributions of mu, delta and kappa opioid receptor mRNA in rat brain. Biochem Biophys Res Commun 1994; 205: 1438–1444.
  • Mansour A, Fox CA, Burke S, Meng F, Thompson RC, Akil H, et al. delta, and kappa opioid receptor mRNA expression in the rat CNS: an in situ hybridization study. J Comp Neurol 1994; 350: 412–438.
  • Mansour A, Khachaturian H, Lewis ME, Akil H, Watson SJ. Anatomy of CNS opioid receptors. Trends Neurosci 1988; 11: 308–314.
  • Okada-Ogawa A, Kurose M, Meng ID. Attenuation of cannabinoid-induced inhibition of medullary dorsal horn neurons by a kappa-opioid receptor antagonist. Brain Res 2010; 1359: 81–89.
  • Stein C, Lang LJ. Peripheral mechanisms of opioid analgesia. Curr Opin Pharmacol 2009; 9: 3–8.
  • Rau KK, Caudle RM, Cooper BY, Johnson RD. Diverse immunocytochemical expression of opioid receptors in electrophysiologically defined cells of rat dorsal root ganglia. J Chem Neuroanat 2005; 29: 255–264.
  • Von Voigtlander PF, Lahti RA, Tang AH, Sethy VH. Biochemical, functional and behavioral evaluation of a series of novel antipsychotic-like agents. Arch Int Pharmacodyn Ther 1983; 266: 23–37.
  • Pfeiffer A, Brantl V, Herz A, Emrich HM. Psychotomimesis mediated by kappa opiate receptors. Science 1986; 233: 774–776.
  • Dykstra LA, Gmerek DE, Winger G, Woods JH. Kappa opioids in rhesus monkeys. I. Diuresis, sedation, analgesia and discriminative stimulus effects. J Pharmacol Exp Ther 1987; 242: 413–420.
  • Liu-Chen LY. Agonist-induced regulation and trafficking of kappa opioid receptors. Life Sci 2004; 75: 511–536.
  • Tominaga M, Ogawa H, Takamori K. Possible roles of epidermal opioid systems in pruritus of atopic dermatitis. J Invest Dermatol 2007; 127: 2228–2235.
  • Cheng B, Liu HW, Fu XB, Sheng ZY, Li JF. Coexistence and upregulation of three types of opioid receptors, mu, delta and kappa, in human hypertrophic scars. Br J Dermatol 2008; 158: 713–720.
  • Salemi S, Aeschlimann A, Reisch N, Jüngel A, Gay RE, Heppner FL, et al. Detection of kappa and delta opioid receptors in skin – outside the nervous system. Biochem Biophys Res Commun 2005; 338: 1012–1017.
  • Bigliardi-Qi M, Gaveriaux-Ruff C, Pfaltz K, Bady P, Baumann T, Rufli T, et al. Deletion of mu- and kappa-opioid receptors in mice changes epidermal hypertrophy, density of peripheral nerve endings, and itch behavior. J Invest Dermatol 2007; 127: 1479–1488.
  • Taneda K, Tominaga M, Negi O, Tengara S, Kamo A, Ogawa H et al. Evaluation of epidermal nerve density and opioid receptor levels in psoriatic itch. Br J Dermatol 2011; 165: 277–284.
  • Messmer D, Hatsukari I, Hitosugi N, Schmidt-Wolf IG, Singhal PC. Morphine reciprocally regulates IL-10 and IL-12 production by monocyte-derived human dendritic cells and enhances T cell activation. Mol Med 2006; 12: 284–290.
  • Finley MJ, Chen X, Bardi G, Davey P, Geller EB, Zhang L, et al. Bi-directional heterologous desensitization between the major HIV-1 co-receptor CXCR4 and the kappa-opioid receptor. J Neuroimmunol 2008; 197: 114–123.
  • Kamei J, Nagase H. Norbinaltorphimine, a selective kappa-opioid receptor antagonist, induces an itch-associated response in mice. Eur J Pharmacol 2001; 418: 141–145.
  • Inan S, Dun NJ, Cowan A. Nalfurafine prevents 5’-guanidinonaltrindole- and compound 48/80-induced spinal c-fos expression and attenuates 5’-guanidinonaltrindole-elicited scratching behavior in mice. Neuroscience 2009; 163: 23–33.
  • Nakao K, Ikeda K, Kurokawa T, Togashi Y, Umeuchi H, Honda T, et al. [Effect of TRK-820, a selective kappa opioid receptor agonist, on scratching behavior in an animal model of atopic dermatitis.] Nihon Shinkei Seishin Yakurigaku Zasshi 2008; 28: 75–83 (in Japanese).
  • Umeuchi H, Togashi Y, Honda T, Nakao K, Okano K, Tanaka T, et al. Involvement of central mu-opioid system in the scratching behavior in mice, and the suppression of it by the activation of kappa-opioid system. Eur J Pharmacol 2003; 477: 29–35.
  • Wang Y, Tang K, Inan S, Siebert D, Holzgrabe U, Lee DY, et al. Comparison of pharmacological activities of three distinct kappa ligands (Salvinorin A, TRK-820 and 3FLB) on kappa opioid receptors in vitro and their antipruritic and antinociceptive activities in vivo. J Pharmacol Exp Ther 2005; 312: 220–230.
  • Umeuchi H, Kawashima Y, Aoki CA, Kurokawa T, Nakao K, Itoh M, et al. Spontaneous scratching behavior in MRL/lpr mice, a possible model for pruritus in autoimmune diseases, and antipruritic activity of a novel kappa-opioid receptor agonist nalfurafine hydrochloride. Eur J Pharmacol 2005; 518: 133–139.
  • Inan S, Cowan A. Nalfurafine, a kappa opioid receptor agonist, inhibits scratching behavior secondary to cholestasis induced by chronic ethynylestradiol injections in rats. Pharmacol Biochem Behav 2006; 85: 39–43.
  • Wakasa Y, Fujiwara A, Umeuchi H, Endoh T, Okano K, Tanaka T, et al. Inhibitory effects of TRK-820 on systemic skin scratching induced by morphine in rhesus monkeys. Life Sci 2004; 75: 2947–2957.
  • Inan S, Cowan A. Kappa opioid agonists suppress chloroquine-induced scratching in mice. Eur J Pharmacol 2004; 502: 233–237.
  • Salemi S, Aeschlimann A, Wollina U, Gay RE, Michel BA, Gay S, et al. Up-regulation of delta-opioid receptors and kappa-opioid receptors in the skin of fibromyalgia patients. Arthritis Rheum 2007; 56: 2464–2466.
  • Lawhorn CD, McNitt JD, Fibuch EE, Joyce JT, Leadley RJ Jr. Epidural morphine with butorphanol for postoperative analgesia after cesarean delivery. Anesth Analg 1991; 72: 53–57.
  • Yeh YC, Lin TF, Lin FS, Wang YP, Lin CJ, Sun WZ. Combination of opioid agonist and agonist-antagonist: patient-controlled analgesia requirement and adverse events among different-ratio morphine and nalbuphine admixtures for postoperative pain. Br J Anaesth 2008; 101: 542–548.
  • Tamdee D, Charuluxananan S, Punjasawadwong Y, Tawichasri C, Patumanond J, Sriprajittichai P. A randomized controlled trial of pentazocine versus ondansetron for the treatment of intrathecal morphine-induced pruritus in patients undergoing cesarean delivery. Anesth Analg 2009; 109: 1606–1611.
  • Nagase H, Hayakawa J, Kawamura K, Kawai K, Takezawa Y, Matsuura H, et al. Discovery of a structurally novel opioid kappa-agonist derived from 4, 5-epoxymorphinan. Chem Pharm Bull (Tokyo) 1998; 46: 366–369.
  • Seki T, Awamura S, Kimura C, Ide S, Sakano K, Minami M, et al. Pharmacological properties of TRK-820 on cloned mu-, delta- and kappa-opioid receptors and nociceptin receptor. J Pharmacol 1999; 376: 159–167.
  • Endoh T, Matsuura H, Tajima A, Izumimoto N, Tajima C, Suzuki T, et al. Potent antinociceptive effects of TRK-820, a novel kappa-opioid receptor agonist. Life Sci 1999; 65: 1685–1694.
  • Endoh T, Tajima A, Izumimoto N, Suzuki T, Saitoh A, Suzuki T, et al. TRK-820, a selective kappa-opioid agonist, produces potent antinociception in cynomolgus monkeys. Jpn J Pharmacol 2001; 85: 282–290.
  • Delmez JA. Efficacy and safety of a new kappa-opioid receptor agonist for the treatment of uremic pruritus. Nat Clin Pract Nephrol 2006; 2: 358–359.
  • Nakao K, Mochizuki H. Nalfurafine hydrochloride: a new drug for the treatment of uremic pruritus in hemodialysis patients. Drugs Today (Barc) 2009; 45: 323–329.
  • Nakao K, Hasebe K, Yoshikawa S, Ikeda K, Hirakata M, Miyamoto Y, et al. [Pharmacological effects of nalfurafine hydrochloride, a kappa-opioid receptor agonist.] Nihon Shinkei Seishin Yakurigaku Zasshi 2010; 30: 185–191 (in Japanese).
  • Tsuji M, Takeda H, Matsumiya T, Nagase H, Yamazaki M, Narita M, et al. A novel kappa-opioid receptor agonist, TRK-820, blocks the development of physical dependence on morphine in mice. Life Sci 2000; 66: 353–358.
  • Delpizzo A. Butorphanol, a new intravenous analgesic: double-blind comparison with morphine sulfate in postoperative patients with moderate or severe pain. Curr Ther Res Clin Exp 1976; 20: 221–232.
  • Heel RC, Brogden RN, Speight TM, Avery GS. Butorphanol: a review of its pharmacological properties and therapeutic efficacy. Drugs 1978; 16: 473–505.
  • Szabova A, Sadhasivam S, Wang Y, Nick TG, Goldschneider K. Comparison of postoperative analgesia with epidural butorphanol/bupivacaine versus fentanyl/bupivacaine following pediatric urological procedures. J Opioid Manag 2010; 6: 401–407.
  • Gunter JB, McAuliffe J, Gregg T, Weidner N, Varughese AM, Sweeney DM. Continuous epidural butorphanol relieves pruritus associated with epidural morphine infusions in children. Paediatr Anaesth 2000; 10: 167–172.
  • Yokoyama Y, Yokoyama T, Nagao Y, Nakagawa T, Magaribuchi T. [Treatment of epidural morphine induced pruritus with butorphanol.] Masui 2009; 58: 178–182 (in Japanese).
  • Sakai T, Fukano T, Sumikawa K. IV butorphanol reduces analgesia but not pruritus or nausea associated with intrathecal morphine. Can J Anaesth 2001; 48: 831–832.
  • Bernstein JE, Grinzi RA. Butorphanol-induced pruritus antagonized by naloxone. J Am Acad Dermatol 1981; 5: 227–228.
  • Dawn AG, Yosipovitch G. Butorphanol for treatment of intractable pruritus. J Am Acad Dermatol 2006; 54: 527–531.
  • Inan S, Cowan A. Reduced kappa-opioid activity in a rat model of cholestasis. Eur J Pharmacol 2005; 518: 182–186.
  • Bergasa NV. Treatment of the pruritus of cholestasis. Curr Treat Options Gastroenterol 2004; 7: 501–508.
  • Lim GJ, Ishiuji Y, Dawn A, Harrison B, Kim do W, Atala A, et al. In vitro and in vivo characterization of a novel liposomal butorphanol formulation for treatment of pruritus. Acta Derm Venereol 2008; 88: 327–330.
  • Olsen JC, McGrath NA, Schwarz DG, Cutcliffe BJ, Stern JL. A double-blind randomized clinical trial evaluating the analgesic efficacy of ketorolac versus butorphanol for patients with suspected biliary colic in the emergency department. Acad Emerg Med 2008; 15: 718–722.