You are on page 1of 3

OPINION ARTICLE

published: 23 September 2013


doi: 10.3389/fphar.2013.00123

Targeting pain and inflammation by peripherally


acting opioids
Christoph Stein*
Department of Anesthesiology and Critical Care Medicine, Charité Campus Benjamin Franklin Freie Universitaet Berlin, Berlin, Germany
*Correspondence: christoph.stein@charite.de
Edited by:
Susan Hua, The University of Newcastle, Australia
Reviewed by:
Peter J. Cabot, The University of Queensland, Australia
Keywords: opioid receptor, opioid peptide, inflammation, pain, analgesia

INTRODUCTION their effects on ion channels, opioids production (Stein and Machelska, 2011;
Opioids can produce potent analgesia attenuate the excitability of peripheral Stein, 2013). Inflammation also leads
by activating opioid receptors out- nociceptor terminals, the propagation of to sprouting of sensory nerve terminals
side the central nervous system, thus action potentials, the release of excitatory and disruption of the perineurial bar-
avoiding centrally mediated unwanted proinflammatory neuropeptides (sub- rier, thus facilitating the access of opi-
effects. Peripheral opioid receptors are stance P, calcitonin gene-related peptide) oid agonists to their receptors (Rittner
expressed in peripheral sensory (dor- from peripheral sensory nerve endings, et al., 2012). Endogenous opioid ligands
sal root ganglion) neurons and can and vasodilatation evoked by stimula- derived from inflammatory cells stimu-
interact with exogenous or endogenous tion of C-fibers. These mechanisms result late recycling of opioid receptors to the
opioid ligands both in animals and in in analgesia and/or anti-inflammatory membrane of sensory neurons, which
humans. Inflammation of peripheral tis- actions (Endres-Becker et al., 2007; Vetter can prevent the development of toler-
sue leads to upregulation of such opioid et al., 2008; Stein and Machelska, 2011; ance to peripherally active opioid ago-
receptors and to local production of Moshourab and Stein, 2012; Nockemann nists (Zöllner et al., 2008). Consistently,
endogenous opioid peptides in immune et al., 2013; Spahn et al., 2013; Stein and clinical studies have indicated a lack of
cells. This article will summarize recent Küchler, 2013). cross-tolerance between peripheral exoge-
mechanistic, preclinical, and clinical nous and endogenous opioids in synovial
findings. PERIPHERAL OPIOID RECEPTORS AND inflammation. All of these mechanisms
TISSUE INJURY can contribute to enhanced antinocicep-
OPIOID RECEPTOR SIGNALING IN Peripheral opioid analgesic effects are par- tive efficacy of opioid agonists in injured
PERIPHERAL SENSORY NEURONS ticularly prominent in inflamed tissue tissue (Stein and Machelska, 2011).
Co-localization and electrophysiological (Kalso et al., 2002; Stein et al., 2003;
studies have confirmed the presence of Vadivelu et al., 2011). Under such con- ENDOGENOUS LIGANDS OF
opioid receptors on C- and A-fibers, on ditions the synthesis and expression of PERIPHERAL OPIOID RECEPTORS
dorsal root ganglion neurons expressing opioid receptors in dorsal root ganglia is Concurrent with the development of
transient receptor potential vanilloid elevated. Subsequently, the axonal trans- inflammation, opioid peptide-producing
subtype-1 (TRPV-1) and G-protein- port and membrane-directed trafficking immune cells are recruited to the site
coupled inwardly rectifying K+ (GIRK) of opioid receptors increases, leading to of injury. The most thoroughly char-
channels, and on fibers expressing isolectin their upregulation on peripheral neu- acterized peptides are β-endorphin and
B4, substance P, and/or calcitonin-gene- ron terminals (Patwardhan et al., 2005; enkephalins deriving from the respec-
related peptide, consistent with the Cayla et al., 2012; Pettinger et al., 2013). tive precursors proopiomelanocortin
phenotype of nociceptors. The activation These events are dependent on neuronal (POMC) and proenkephalin. Transcripts
of such opioid receptors results in inhibi- electrical activity, cytokines, and nerve and peptides derived from POMC and
tion of high-voltage activated Ca++ - and growth factor from the damaged tissue. In proenkephalin, as well as the prohor-
enhancement of GIRK-currents. These mechanical nerve injury leading to neuro- mone convertases PC1/3 and PC2,
effects are mediated by G-proteins (Gi pathic pain, opioid receptors accumulate necessary for their posttranslational pro-
and/or Go ). In addition, opioids—via proximal and distal to the lesion, indi- cessing, were detected in such cells. The
inhibition of adenylyl cyclase—suppress cating anterograde and retrograde trans- expression of immune-derived opioids
tetrodotoxin-resistant Na+ -, TRPV1- and port (Labuz et al., 2009). Inflammatory is stimulated by viruses, endotoxins,
other non-selective cation currents stim- milieu (low pH, prostanoids, bradykinin) cytokines, corticotropin releasing hor-
ulated by inflammatory agents, which can augment opioid receptor function mone (CRH) and adrenergic agonists.
may account for the notable efficacy e.g., by more efficient G-protein cou- In painful tissue inflammation and neu-
of peripheral opioids in inflammatory pling and inhibition of elevated neu- ropathy, POMC mRNA, β-endorphin,
and neuropathic pain. Consistent with ronal cyclic adenosine monophosphate met-enkephalin, and dynorphin are

www.frontiersin.org September 2013 | Volume 4 | Article 123 | 1


Stein Peripherally acting opioids

detectable in circulating cells and lymph with minimal capability to cross the lack of clinical studies in this area at
nodes, and are upregulated in resident blood-brain-barrier. Among the first present.
lymphocytes, monocytes/macrophages, compounds were the mu-agonist lop-
and granulocytes. Circulating opioid- eramide (known as an antidiarrheal CLINICAL STUDIES ON PERIPHERAL
containing leukocytes migrate to injured drug) and the kappa-agonist asimadoline. OPIOID ANALGESICS
tissue attracted by adhesion molecules, Peripheral restriction was also achieved The most extensively examined clinical
chemokines, and neurokinins. In inflamed with glucuronidation, arylacetamide application is the intraarticular injection
tissue, opioid-containing leukocytes, vas- (ADL 10-0101), morphinan-based (TRK- of morphine. Both in human and vet-
cular P-selectin, ICAM-1, and PECAM-1 820, HS-731), triazaspiro (DiPOA) and erinary medicine, numerous controlled
are simultaneously upregulated. Blocking peptidic compounds (DALDA, FE200665, clinical studies have demonstrated dose-
chemokines, selectins, or ICAM-1 reduces CR845). While earlier attempts to demon- dependent and peripherally mediated
the extravasation of opioid-containing strate peripheral opioid analgesia in reduction of pain and/or supplemental
cells and increases inflammatory and normal tissue failed, they were much more analgesic consumption without signif-
neuropathic pain. Consistently, immuno- successful in models of pathological pain icant side effects (Kalso et al., 2002;
suppression can exacerbate pain (Labuz (Stein, 1993). For example, in subcuta- Stein, 2013). Intraarticular morphine
et al., 2009; Stein and Machelska, 2011; neous inflammation the local injection of is effective in acute (postoperative) and
Busch-Dienstfertig et al., 2012). low, systemically inactive doses of mu-, chronic (arthritic) pain, its effect is simi-
Stimuli such as environmental stress, delta-, and kappa-agonists produces dose- lar to intraarticular local anesthetics and
noradrenaline, CRH, interleukin-1β, dependent and opioid receptor-specific steroids, and it is long lasting, possibly
chemokines, or mycobacteria can elicit antinociception. Such effects were also due to anti-inflammatory activity. Locally
opioid peptide release from immune cells shown in models of nerve damage, vis- applied opioids were also effective in den-
via specific receptors and the regulated ceral, thermal, cancer and bone pain (Stein tal pain, skin ulcers, corneal abrasions
secretory pathway. Depending on the cell and Machelska, 2011). and visceral pain (Sawynok, 2003; Farley,
type and agent, intracellular Ca++ release 2011; Vadivelu et al., 2011). Some stud-
from endoplasmic reticulum or extracellu- EFFECTS ON INFLAMMATION ies found no peripheral effects of opioids,
lar Ca++ is required. In vivo, the secreted Inflammation contributes to many diverse e.g., after injection into the non-inflamed
opioid peptides bind to opioid receptors disorders such as trauma, arthritis, environment along nerve trunks (Picard
on sensory neurons and elicit analgesia neuropathy, fibromyalgia, endometrio- et al., 1997). The latter observation sug-
in injured tissue and neuropathy (Labuz sis, diabetes, cancer, and chronic pain. gests that intraaxonal opioid receptors are
et al., 2009; Rittner et al., 2009). Not only Therapeutic inhibition of inflammation “in transit,” and not available as functional
stimulated but also tonic release of opioids is indicated when it becomes dysregu- receptors at the membrane. Peripherally
from immune cells decreases pain in ani- lated, chronic, recurrent or inappropriate. restricted opioids are under investigation
mals (Rittner et al., 2009) and in humans However, standard treatments such as for human use (morphine-6-glucuronide,
(Stein et al., 1993). Thus, the develop- steroids, non-steroidal anti-inflammatory CR845), and were shown to reduce post-
ment of inflammatory and neuropathic drugs (NSAIDs), and disease-modifying operative and visceral pain with similar
pain is counteracted by immune cells drugs have severe side effects (ulcers, efficacy as morphine but limited central
producing and secreting opioid peptides. bleeding, myocardial infarction, stroke, side-effects (Dahan et al., 2008; Binning
Gene therapeutic approaches are aiming to infections) (Trelle et al., 2011) and bio- et al., 2011; Stein and Machelska, 2011).
increase the production of opioid peptides logical anti-inflammatory treatments such
and receptors in inflammatory cells and as inhibitors of tumor necrosis factor- SUMMARY
peripheral sensory neurons, respectively α or of Janus kinases can only be used Opioids can reduce pain and inflamma-
(Stein and Machelska, 2011; Raja, 2012). in a limited number of patients due to tion by activating opioid receptors outside
Preventing the extracellular degradation of their prohibitive cost, parenteral formu- the central nervous system. Inflammation
endogenous opioid peptides by peptidase lation and risk for infection and tumor of peripheral tissue leads to upregula-
inhibitors as well as nanocarrier-directed induction. A large number of in vitro tion of opioid receptors on peripheral
transport of opioids have been shown and animal investigations have produced sensory neurons and to local produc-
to diminish inflammatory pain (Roques evidence that peripherally active opioids tion of endogenous opioid peptides in
et al., 2012; Schreiter et al., 2012; Hua and can reduce release of proinflammatory immune cells. Future aims in drug devel-
Cabot, 2013). neuropeptides, cytokines, plasma extrava- opment include the design of peripher-
sation, vasodilation, immune mediators, ally restricted opioid agonists, selective
PRECLINICAL STUDIES ON expression of adhesion molecules and targeting of opioids to sites of painful
PERIPHERAL OPIOID ANALGESICS tissue destruction (Stein and Küchler, injury and the augmentation of periph-
This basic research has stimulated the 2012). In contrast to currently avail- eral ligand and receptor synthesis, e.g.,
development of novel opioid ligands act- able anti-inflammatory agents, opioids by gene therapy. The ultimate goal is to
ing exclusively in the periphery without have no demonstrated organ toxicity, avoid detrimental side effects of currently
central side-effects. A common approach making them interesting candidates for available opioid and nonopioid drugs
is the use of hydrophilic compounds drug development. However, there is a such as apnoea, cognitive impairment,

Frontiers in Pharmacology | Neuropharmacology September 2013 | Volume 4 | Article 123 | 2


Stein Peripherally acting opioids

addiction, gastrointestinal bleeding, and inflammation. J. Neurophysiol. 108, 2827–2836. Stein, C. (2013). Towards safer and more
thromboembolic complications. doi: 10.1152/jn.00082.2012 effective analgesia. Vet. J. 196, 6–7. doi:
Nockemann, D., Rouault, M., Labuz, D., Hublitz, P., 10.1016/j.tvjl.2012.09.013
ACKNOWLEDGMENTS McKnelly, K., Reis, F. C., et al. (2013). The K Stein, C., Hassan, A. H. S., Lehrberger, K., Giefing,
channel GIRK2 is both necessary and sufficient J., and Yassouridis, A. (1993). Local analgesic
Supported by Bundesministerium für for peripheral opioid-mediated analgesia. EMBO effect of endogenous opioid peptides. Lancet
Bildung und Forschung (VIP0272/AZ Mol. Med. 5, 1263–1277. doi: 10.1002/emmm. 342, 321–324. doi: 10.1016/0140-6736(93)
03V0364; 0316177B/C1). 201201980 91471-W
Patwardhan, A. M., Berg, K. A., Akopain, A. N., Stein, C., and Küchler, S. (2012). Non-analgesic
REFERENCES Jeske, N. A., Gamper, N., Clarke, W. P., et al. effects of opioids: peripheral opioid effects on
Binning, A. R., Przesmycki, K., Sowinski, P., (2005). Bradykinin-induced functional com- inflammation and wound healing. Curr. Pharm.
Morrison, L. M., Smith, T. W., Marcus, petence and trafficking of the delta-opioid Des. 18, 6053–6069. doi: 10.2174/13816121280
P., et al. (2011). A randomised controlled receptor in trigeminal nociceptors. J. Neurosci. 3582513
trial on the efficacy and side-effect profile 25, 8825–8832. doi: 10.1523/JNEUROSCI. Stein, C., and Küchler, S. (2013). Targeting inflam-
(nausea/vomiting/sedation) of morphine-6- 0160-05.2005 mation and wound healing by opioids. Trends
glucuronide versus morphine for post-operative Pettinger, L., Gigout, S., Linley, J. E., and Gamper, Pharmacol. Sci. 34, 303–312. doi: 10.1016/j.tips.
pain relief after major abdominal surgery. Eur. N. (2013). Bradykinin controls pool size of sen- 2013.03.006
J. Pain 15, 402–408. doi: 10.1016/j.ejpain.2010. sory neurons expressing functional delta-opioid Stein, C., and Machelska, H. (2011). Modulation
09.007 receptors. J. Neurosci. 33, 10762–10771. doi: of peripheral sensory neurons by the immune
Busch-Dienstfertig, M., Labuz, D., Wolfram, 10.1523/JNEUROSCI.0123-13.2013 system: implications for pain therapy. Pharmacol.
T., Vogel, N. N., and Stein, C. (2012). Picard, P. R., Tramer, M. R., McQuay, H. J., and Rev. 63, 860–881. doi: 10.1124/pr.110.
JAK-STAT1/3-induced expression of signal Moore, R. A. (1997). Analgesic efficacy of periph- 003145
sequence-encoding proopiomelanocortin mRNA eral opioids (all except intra-articular): a qualita- Stein, C., Schäfer, M., and Machelska, H. (2003).
in lymphocytes reduces inflammatory pain tive systematic review of randomised controlled Attacking pain at its source: new perspec-
in rats. Mol. Pain 8, 83. doi: 10.1186/1744- trials. Pain 72, 309–318. doi: 10.1016/S0304- tives on opioids. Nat. Med. 9, 1003–1008. doi:
8069-8-83 3959(97)00040-7 10.1038/nm908
Cayla, C., Labuz, D., Machelska, H., Bader, M., Raja, S. N. (2012). Modulating pain in the periph- Trelle, S., Reichenbach, S., Wandel, S., Hildebrand,
Schäfer, M., and Stein, C. (2012). Impaired ery: gene-based therapies to enhance periph- P., Tschannen, B., Villiger, P. M., et al.
nociception and peripheral opioid antinoci- eral opioid analgesia: bonica lecture, ASRA (2011). Cardiovascular safety of non-steroidal
ception in mice lacking both kinin B1 and B2 (2010). Reg. Anesth. Pain Med. 37, 210–214. doi: anti-inflammatory drugs: network meta-
receptors. Anesthesiology 116, 448–457. doi: 10.1097/AAP.0b013e31823b145f analysis. BMJ 342:c7086. doi: 10.1136/
10.1097/ALN.0b013e318242b2ea Rittner, H. L., Amasheh, S., Moshourab, R., bmj.c7086
Dahan, A., van Dorp, E., Smith, T., and Yassen, A. Hackel, D., Yamdeu, R. S., Mousa, S. A., Vadivelu, N., Mitra, S., and Hines, R. L.
(2008). Morphine-6-glucuronide (M6G) for post- et al. (2012). Modulation of tight junction (2011). Peripheral opioid receptor ago-
operative pain relief. Eur. J. Pain 12, 403–411. doi: proteins in the perineurium to facilitate nists for analgesia: a comprehensive review.
10.1016/j.ejpain.2007.07.009 peripheral opioid analgesia. Anesthesiology J. Opioid Manag. 7, 55–68. doi: 10.5055/jom.
Endres-Becker, J., Heppenstall, P. A., Mousa, S. A., 116, 1323–1334. doi: 10.1097/ALN.0b013e318 2011.0049
Labuz, D., Oksche, A., Schäfer, M., et al. (2007). 256eeeb Vetter, I., Cheng, W., Peiris, M., Wyse, B. D., Roberts-
Mu-opioid receptor activation modulates transient Rittner, H. L., Hackel, D., Voigt, P., Mousa, S., Thomson, S. J., Zheng, J., et al. (2008). Rapid,
receptor potential vanilloid 1 (TRPV1) currents in Stolz, A., Labuz, D., et al. (2009). Mycobacteria opioid-sensitive mechanisms involved in transient
sensory neurons in a model of inflammatory pain. attenuate nociceptive responses by formyl pep- receptor potential vanilloid 1 sensitization. J. Biol.
Mol. Pharmacol. 71, 12–18. doi: 10.1124/mol.106. tide receptor triggered opioid peptide release Chem. 283, 19540–19550. doi: 10.1074/jbc.M70
026740 from neutrophils. PLoS Pathog. 5:e1000362. doi: 7865200
Farley, P. (2011). Should topical opioid anal- 10.1371/journal.ppat.1000362 Zöllner, C., Mousa, S. A., Fischer, O., Rittner, H. L.,
gesics be regarded as effective and safe Roques, B. P., Fournie-Zaluski, M. C., and Wurm, Shaqura, M., Brack, A., et al. (2008). Chronic
when applied to chronic cutaneous lesions. M. (2012). Inhibiting the breakdown of endoge- morphine use does not induce peripheral tol-
J. Pharm. Pharmacol. 63, 747–756. doi: nous opioids and cannabinoids to alleviate erance in a rat model of inflammatory pain.
10.1111/j.2042-7158.2011.01252.x pain. Nat. Rev. Drug Discov. 11, 292–310. doi: J. Clin. Invest. 118, 1065–1073. doi: 10.1172/
Hua, S., and Cabot, P. J. (2013). Targeted nanopar- 10.1038/nrd3673 JCI25911
ticles that mimic immune cells in pain con- Sawynok, J. (2003). Topical and peripherally act-
trol inducing analgesic and anti-inflammatory ing analgesics. Pharmacol. Rev. 55, 1–20. doi: Received: 05 August 2013; accepted: 05 September 2013;
actions: a potential novel treatment of acute 10.1124/pr.55.1.1 published online: 23 September 2013.
and chronic pain condition. Pain Physician 16, Schreiter, A., Gore, C., Labuz, D., Fournie-Zaluski, Citation: Stein C (2013) Targeting pain and
E199–E216. M. C., Roques, B. P., Stein, C., et al. (2012). inflammation by peripherally acting opioids. Front.
Kalso, E., Smith, L., McQuay, H. J., Moore, R. Pain inhibition by blocking leukocytic and neu- Pharmacol. 4:123. doi: 10.3389/fphar.2013.00123
A. (2002). No pain, no gain: clinical excellence ronal opioid peptidases in peripheral inflamed tis- This article was submitted to Neuropharmacology, a
and scientific rigour - lessons learned from IA sue. FASEB J. 26, 5161–5171. doi: 10.1096/fj.12- section of the journal Frontiers in Pharmacology.
morphine. Pain 98, 269–275. doi: 10.1016/S0304- 208678 Copyright © 2013 Stein. This is an open-access
3959(02)00019-2 Spahn, V., Fischer, O., Endres-Becker, J., Schäfer, article distributed under the terms of the Creative
Labuz, D., Schmidt, Y., Schreiter, A., Rittner, H. L., M., Stein, C., and Zöllner, C. (2013). Opioid Commons Attribution License (CC BY). The use, dis-
Mousa, S. A., and Machelska, H. (2009). Immune withdrawal increases transient receptor poten- tribution or reproduction in other forums is permit-
cell-derived opioids protect against neuropathic tial vanilloid 1 activity in a protein kinase ted, provided the original author(s) or licensor are
pain in mice. J. Clin. Invest. 119, 278–286. doi: A-dependent manner. Pain 154, 598–608. doi: credited and that the original publication in this
10.1172/JCI36246 10.1016/j.pain.2012.12.026 journal is cited, in accordance with accepted aca-
Moshourab, R., and Stein, C. (2012). Fentanyl Stein, C. (1993). Peripheral mechanisms of opi- demic practice. No use, distribution or reproduc-
decreases discharges of C and A nociceptors to oid analgesia. Anesth. Analg. 76, 182–191. doi: tion is permitted which does not comply with these
suprathreshold mechanical stimulation in chronic 10.1213/00000539-199301000-00031 terms.

www.frontiersin.org September 2013 | Volume 4 | Article 123 | 3

You might also like