You are on page 1of 43

The Oxford Handbook of the Neurobiology of Pain

John N. Wood (ed.)

https://doi.org/10.1093/oxfordhb/9780190860509.001.0001
Published: 2018 Online ISBN: 9780190860530 Print ISBN: 9780190860509

CHAPTER

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


25 Opioids and Pain 
Christoph Stein, Claire Gaveriaux-Ru

https://doi.org/10.1093/oxfordhb/9780190860509.013.9 Pages 728–769


Published: 10 July 2018

Abstract
This article reviews basic and clinical concepts on pain and mechanisms underlying opioid analgesia. It
describes the structure, function, and cellular signaling of opioid receptors; endogenous and
exogenous opioid receptor ligands; as well as the central and peripheral sites of opioid actions. The
article also presents novel opioid-based therapeutic strategies, developed from recently gained
knowledge on opioid receptor structures and signaling, pathological pain situations, and mutant
mouse lines, aimed at the reduction of side e ects such as respiratory depression, constipation,
sedation, tolerance, or opioid-induced hyperalgesia. Lastly, the article addresses clinical problems
associated with opioid use, particularly the long-term use of opioid analgesics in chronic pain.

Keywords: Opioid receptors, analgesia, ligands, peripheral, respiratory depression, sedation, constipation,
tolerance, hyperalgesia
Subject: Molecular and Cellular Systems, Neuroscience
Series: Oxford Handbooks
Collection: Oxford Handbooks Online

Acute and Chronic Pain

Basic Concepts
Pain may be roughly divided into two broad categories: physiological and pathological pain. Physiological
(acute, nociceptive) pain is an essential early warning sign that usually elicits re ex withdrawal and thereby
promotes survival by protecting the organism from (further) injury. In contrast, pathological (e.g., chronic
neuropathic) pain is an expression of the maladaptive operation of the nervous system; it is pain as a
disease (Flor & Diers, 2007; Fordyce, Fowler, & DeLateur, 1968; Stein, 2013a).
Excitatory Mechanisms
Physiological pain is mediated by a sensory system consisting of primary a erent neurons, spinal
interneurons, ascending tracts, and supraspinal areas. Trigeminal and dorsal root ganglia (DRG) give rise to
high-threshold Aδ– and C- bers innervating peripheral tissues (skin, muscles, joints, viscera). These
specialized primary a erent neurons (nociceptors) transduce noxious stimuli into action potentials and
conduct them to the dorsal horn of the spinal cord. When peripheral tissue is damaged, primary a erent
neurons are sensitized and/or directly activated by thermal, mechanical, and/or chemical stimuli. Examples
are sympathetic amines, adenosine triphosphate (ATP), neuropeptides, nerve growth factor, prostanoids,
bradykinin, proin ammatory cytokines, and protons (Table 25.1) (Basbaum, Bautista, Scherrer, & Julius,

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


2009). Many of these agents lead to opening of cation channels (e.g., TRPV1, P2X3) in the neuronal
membrane. This produces inward depolarizing currents and subsequent action potentials that are then
conducted along the sensory axon to the dorsal horn of the spinal cord. Thereafter, these impulses are
transmitted to spinal neurons, brainstem, thalamus, and cortex. Repeated nociceptor stimulation can
p. 730 sensitize peripheral and central neurons

p. 731
(activity-dependent plasticity, or “wind-up”). This can be sustained by changes in the expression of genes
coding for various neuropeptides, transmitters, ion channels, receptors, and signaling molecules
(transcription-dependent plasticity) in peripheral and central neurons (Baron, Hans, & Dickenson, 2013;
Basbaum et al., 2009). Both induction and maintenance of central sensitization are critically dependent on
the peripheral drive by nociceptors, indicating that therapeutic interventions targeting such neurons may
be particularly e ective, even in chronic pain syndromes (Baron et al., 2013; Richards & McMahon, 2013).
Table 25.1 pH Values in Inflamed Tissues Measured in Vivo/ex Vivo

Reference Species, tissue Lowest


pH

(Häbler, 1929) Human, abscess 5.4

(Koldajew & Altschuler, 1930) Guinea pig, intraperitoneal bacterial inoculation 5.6
Mouse, s.c., i.p. bacterial inoculation 5.8

(Menkin, 1934) Dog, turpentine-induced pleural exudate 6.6

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


(Voegtlin, 1935) Rat, implanted tumors 6.82

(Menkin & Warner, 1937) Dog, turpentine-induced pleural exudate 6.5

(Meyer, Kammerling, & et al., 1948) Human, malignant tumors, inflamed tissues 5.44

(Revici, Stoopen, Frenk, & Ravich, 1949) Human, malignant tumor 5.7

(Menkin, 1956) Dog, turpentine-induced pleural exudate 6.0

(Jebens & Monk-Jones, 1959) Human, osteoarthritis, joint injury, synovial fluid 6.5

(Pampus, 1963) Human, astrocytoma 5.85

(Ashby, 1966) Human, melanoma 6.4

(Cummings & Nordby, 1966) Human, rheumatoid arthritis synovial fluid 7.08

(Goldie & Nachemson, 1969) Human, rheumatoid arthritis synovial fluid 6.0

(Goldie & Nachemson, 1970) Human, rheumatoid arthritis synovial fluid 6.4

(Falchuk, Goetzl, & Kulka, 1970) Human, rheumatoid arthritis 6.84

(Treuha & McCarty, 1971) Human, arthritis 6.60

(Hutchins & Sheldon, 1972) Rabbit, diabetic skin wounds 6.9

(Silver, 1975) Rabbit, brain, wounds, ischemia 5.0

(Jacobus, Taylor, Hollis, & Nunnally, 1977) Rat, ischemic heart, intracellular 5.7

(Levine & Kelly, 1978) Rat, seminiferous tubules and epididymis 6.57+0.08

(Vaupel, Frinak, & Bicher, 1981) Mouse mammary carcinoma 5.8

(Farr, Garvey, Bold, Kendall, & Bacon, 1985) Human, rheumatoid and osteoarthritis synovial fluid 6.85

(Punnia-Moorthy, 1987) Rat, air pouch granuloma induced by carrageenan, dextran, 6.87
Staph. aureus

(Pan, Hamm, Rothman, & Shulman, 1988) Human, exercised muscle, intracellular pH 6.1

(Hood, Schubert, Keller, & Muller, 1988) Human, exercised muscle, calculated intracellular pH 6.31+0.09

(Geborek, Saxne, Pettersson, & Wollheim, Human, rheumatoid arthritis synovial fluid 7.03
1989)

(Tulamo, Bramlage, & Gabel, 1989) Horse, Staph. aureus-induced arthritis, synovial fluid 6.2

(Newell, Franchi, Pouyssegur, & Tannock, Nude mouse; implanted tumors 6.65
1993)
(Simmen & Blaser, 1993) Human, abdominal abscess 6.0

(Gillies, Liu, & Bhujwalla, 1994) Mouse, implanted tumor 6.66

(Alfaro et al., 1996) Rat, carrageenan inflammation, aspirated 6.94

(Issberner, Reeh, & Steen, 1996) Human, exercised muscle, intracutaneous pH 6.67

(Stubbs, McSheehy, & Gri iths, 1999) Rat, mouse, implanted tumors 6.3

(Ojugo et al., 1999) Mouse, implanted tumors 6.0

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


(Andersson, Lexmuller, Johansson, & Rat, bovine serum albumin (BSA)-induced arthritis 5.66
Ekstrom, 1999)

(Woo, Park, Subieta, & Brennan, 2004) Rat, plantar/ gastrocnemius incision 6.54+0.12

(Gallagher et al., 2008) Mouse, implanted subcutaneous lymphoma 6.0

(Spahn et al., 2017) Rat, Freundʼs adjuvant paw inflammation; 6.8


paw incision 7.02

(Gonzalez-Rodriguez et al., 2017) Rat, Freundʼs adjuvant paw inflammation 6.82

Adapted from Stein, 2018.

Abbreviations: s.c., subcutaneous; i.p., intraperitoneal; BSA, bovine serum albumin; NMR, nuclear magnetic resonance; MRS,
magnetic resonance spectroscopy, MRI, magnetic resonance imaging; 3-APP, 3-aminopropylphosphonate.

Inhibitory Mechanisms
Concurrent with such excitatory events, powerful endogenous mechanisms counteracting pain unfold. This
was initially proposed in the “gate control theory of pain” of 1965 and has since been corroborated and
expanded by experimental data in the central nervous system (CNS) and in the periphery. In 1990, a
“peripheral gate” was discovered at the source of pain generation by demonstrating that immune cell–
derived opioid peptides can block the excitation of nociceptors carrying opioid receptors within injured
tissue (Figure 25.1) (Stein et al., 1990). This represented the rst example of many subsequently described
neuro-immune interactions relevant to pain (Machelska, 2011; Stein, 1995; Stein & Machelska, 2011). In the
spinal cord, pain inhibition is mediated by the release of opioid peptides, gamma-amino-butyric acid
p. 732 (GABA), or glycine. During ongoing nociceptive stimulation, spinal interneurons upregulate gene
expression and production of opioid peptides (Herz, Millan, & Stein, 1989). Powerful descending inhibitory
pathways from the brainstem also become active by operating through noradrenergic, serotonergic, and
opioid systems (Basbaum et al., 2009; Schumacher, Basbaum, & Naidu, 2015). The supraspinal integration
of signals from excitatory and inhibitory neurotransmitters, and cognitive, emotional, and environmental
factors eventually results in the central perception of pain.
Figure 25.1.

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Antinociceptive mechanisms within peripheral injured tissue. Opioid peptide-containing circulating leukocytes extravasate upon
activation of adhesion molecules and chemotaxis by chemokines. Subsequently, these leukocytes are stimulated by stress or
releasing agents to secrete opioid peptides. For example, corticotropin-releasing factor (CRF), interleukin-1β (IL-1) and
noradrenaline (NA; released from postganglionic sympathetic neurons) can elicit opioid release by activating their respective
CRF receptors (CRFR), IL-1 receptors (IL-1R), and adrenergic receptors (AR) on leukocytes. Exogenous opioids (EO) or endogenous
opioid peptides (OP; green triangles) bind to opioid receptors (OR) that are synthesized in dorsal root ganglia and transported
along intra-axonal microtubules to peripheral (and central) terminals of sensory neurons. The subsequent inhibition of ion
++
channels (e.g., TRPV1, Ca ) and of substance P (sP) release results in anti-nociceptive e ects.

Adapted with permission from C. Stein, Opioid Receptors, Annual Review of Medicine, 67, 433–451, © 2016, Annual Reviews.

Translation of Basic Research into Clinical Applications


Basic research on pain continues at a rapid pace, but translation into clinical applications has been di cult
p. 733 (Richards & McMahon, 2013; Yekkirala, Roberson, Bean, & Woolf, 2017). Many obstacles have been
discussed, including over-interpretation of data, reporting-bias towards neglecting negative results,
inadequate animal models, awed study design, and genetic and species di erences (Berge, 2011; Mogil,
Davis, & Derbyshire, 2010; Richards & McMahon, 2013; Yekkirala et al., 2017). Notwithstanding these issues,
animal studies are indispensable, continue to be improved, and have successfully predicted adverse side
e ects of drug candidates (Berge, 2011; Mogil et al., 2010). To better represent the complex dimensions of
human pain, natural models and the social transfer of pain have been investigated recently (Klinck et al.,
2017; Smith, Hostetler, Heinricher, & Ryabinin, 2016). For ethical reasons, many models are restricted to
days or weeks, while human chronic pain can last for months or years. Therefore, animal models may be
more cautiously termed as re ecting “persistent” pain (Berge, 2011; Yekkirala et al., 2017).

Brain imaging is another area of intense research. Numerous studies have investigated changes in patients
with various pain syndromes, but they have not provided reproducible ndings speci c for a disease or a
pathophysiological basis for individual syndromes (Mogil et al., 2010). Some studies suggested that the
imaged activity may re ect salience rather than intensity of pain (Legrain, Iannetti, Plaghki, & Mouraux,
2011). Imaging of opioid mechanisms in the human brain has been limited mostly to single-dose studies in
healthy volunteers and has not substantially advanced our understanding of pain relief or opioid use in
patients (Lee, Wanigasekera, & Tracey, 2012). Neuroimaging can only detect alterations associated with
nociceptive processes, whereas clinical pain encompasses a much more complex subjective experience that
critically relies on self-evaluation. Thus, although recent data have provided valuable information on pain
neurophysiology, current imaging techniques cannot yet provide an objective proxy, biomarker, or
predictor for clinical pain (Davis et al., 2017; Smith et al., 2017).

Genetics is another budding scienti c eld. However, with the possible exception of the metabolic enzyme
CYP2D6, pharmacogenetics is not expected to serve as a guide to individualized (“personalized”) clinical
pain therapy any time soon (Chidambaran, Sadhasivam, & Mahmoud, 2017; Drewes et al., 2013; Kringel et
al., 2017; Matic, de Wildt, Tibboel, & van Schaik, 2017; Mogil et al., 2010; Mura et al., 2013; Roberts et al.,
2012; Walter, Doehring, Oertel, & Lötsch, 2013).

Clinical Concepts: Definitions and Prevalence

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


The International Association for the Study of Pain (IASP) de nes pain as “an unpleasant sensory and
emotional experience associated with actual or potential tissue damage, or described in terms of such
damage” (Loeser & Treede, 2008). Nociception is neurophysiological activity in peripheral sensory neurons
(nociceptors) and higher nociceptive pathways and is de ned by the IASP as the “neural process of encoding
noxious stimuli.” Nociception is not synonymous with pain. Pain is always a psychological state, even
p. 734 though it often has a proximate physical cause. When the intricate balance between biological
(neuronal), psychological (e.g., learning, memory, distraction), and social (e.g., attention, reward) factors
becomes disturbed, chronic pain can develop (Flor & Diers, 2007; Fordyce, 1992; Fordyce et al., 1968; Stein,
2013a). Chronic pain has enormous socioeconomic costs due to health care, disability compensation, lost
workdays, and related expenses (Dzau & Pizzo, 2014; Leadley, Armstrong, Lee, Allen, & Kleijnen, 2012).

There is a tradition of distinguishing between non-malignant (e.g., neuropathic, musculoskeletal,


in ammatory) and malignant (related to cancer and its treatment) chronic pain. Non-malignant chronic
pain is frequently classi ed into in ammatory (e.g., arthritic), musculoskeletal (e.g., low back pain),
headaches, and neuropathic pain (e.g., post-herpetic neuralgia, phantom pain, complex regional pain
syndrome, diabetic neuropathy, HIV neuropathy) (Baron, 2006). Cancer pain can originate from the
invasion of the tumor into tissues innervated by primary a erent neurons (e.g., pleura, peritoneum) or
directly into peripheral nerve plexus. In the latter case, neuropathic symptoms may be predominant.

Bio-psycho-social Concept
Both cancer and non-cancer patients with chronic pain have in common the complex in uences of
biological (tissue damage), cognitive (memory, expectations), emotional (anxiety, depression), and
environmental factors (reinforcement, conditioning). Pain behaviors such as limping, medication intake, or
avoidance of activity are all subject to operant conditioning; that is, they respond to reward and
punishment. For example, pain behaviors may be positively reinforced by attention from a spouse or
healthcare provider (e.g., by inadequate use of medications). Conversely, such behaviors can be
extinguished when they are disregarded or when incremental activity is reinforced by social attention and
praise (Fordyce et al., 1968). The interplay between biological, psychological, and social factors results in
the persistence of pain and illness behaviors (Flor & Diers, 2007; Fordyce et al., 1968). Besides possible
long-term neuronal sensitization, this concept helps us understand why chronic pain may exist without
obvious physical cause.
Pain Management
The treatment of both acute (e.g., postoperative) and chronic pain remains a major challenge in clinical
medicine and public health (Chou et al., 2016; Dzau & Pizzo, 2014; Richards & McMahon, 2013). One
component of pain therapy is the use of analgesic drugs. They interfere with the generation and/or
transmission of impulses in the peripheral and/or CNS (nociception) (Yekkirala et al., 2017). Drugs currently
used in clinical pain treatment include opioids, nonsteroidal anti-in ammatory drugs (NSAIDs),
serotonergic compounds, antiepileptics, and antidepressants (Stein & Kopf, 2015). Placebo treatments have
also shown impressive analgesic e ects, mediated by opioid and non-opioid mechanisms (Carlino, Pollo, &
Benedetti, 2011). In chronic pain, treating only nociception is obviously insu cient. A bio-psycho-social

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


p. 735 approach addresses physical, psychological, and social skills and underscores the patients’ active
responsibility to regain control over their life by improving their function and well-being (Engel, von Kor ,
& Katon, 1996; Flor & Diers, 2007; Gatchel & Okifuji, 2006; Stein & Kopf, 2015).

Opioids

The opioid receptor family contains three pharmacologically distinct receptors, named mu-, delta-, and
kappa-receptors (MOR, DOR, KOR), which are encoded by the genes OPRM1, OPRD1 and OPRK1. Opioid
receptors (OR) are seven transmembrane G protein–coupled receptors (GPCR) that are the physiological
targets of endogenous opioid peptides (Figure 25.2). ORs are expressed on neurons and many other cell
types, including neuroendocrine, immune, heart, and skin cells (Stein & Machelska, 2011). Cloning and
sequence analysis of OR genes have demonstrated high homology (Law, Reggio, & Loh, 2013; Wei & Loh,
2011). Other receptors (sigma, orphaninFQ/nociception, epsilon) are no longer considered classical ORs.

Figure 25.2.

The opioid system is composed of the three opioid receptors mu (MOR), delta (DOR), and kappa (KOR), and of the endogenous
opioid peptides endorphins, enkephalins, and dynorphins. Opioid peptides activate opioid receptors with low receptor type
selectivity, endomorphins and endogenous morphine show selectivity for MOR. Exogenous opioids (morphine, fentanyl,
oxycodone) act selectively on MOR to elicit analgesia and other e ects.
p. 736 Opioid Receptor Gene Variants
The mu-opioid receptor gene OPRM1 was among the rst genes to be screened for functional relevance with
regard to analgesia. The human single-nucleotide polymorphism (SNP) OPRM1 118 A>G is the most
thoroughly investigated candidate to date. In vitro biochemical and molecular assays indicated altered
binding a nity, signal transduction, and expression. Therefore, it was assumed that this might underlie
occasionally diminished opioid e cacy in patients. However, as emerged from meta-analyses, these
ndings translate only into very small clinical e ects, such as slightly higher opioid-dosing requirements
for acute pain, but they do not a ect chronic pain or opioid side e ects. Thus, this SNP appears to be
without major clinical relevance as a solitary variant, and it is not a useful biomarker on which therapeutic

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


decisions can be based (Mura et al., 2013; Walter et al., 2013). This is probably due to the complexity of pain,
which is known to be regulated by more than 400 genes with functionally opposite variants that are
concomitantly present (Walter et al., 2013). Nonetheless, e orts continue to nd other genetic variants as
potential biomarkers by use of bioinformatics and next-generation sequencing techniques (Bruehl et al.,
2013; Busch-Dienstfertig, Roth, & Stein, 2013; Hauser et al., 2018; Kringel et al., 2017)

Endogenous Ligands
Endogenous opioid peptides are derived from the precursors proopiomelanocortin (encoding beta-
endorphin), proenkephalin (encoding Met-enkephalin and Leu-enkephalin), and prodynorphin (encoding
dynorphins). These peptides contain the common Tyr-Gly-Gly-Phe-Met/Leu sequence at their amino
terminals, known as the “opioid motif.” Beta-endorphin and the enkephalins are anti-nociceptive agents
acting at MOR and DOR. Dynorphins can elicit both pro- and anti-nociceptive e ects via N-methyl-D-
aspartate (NMDA) receptors, bradykinin receptors, and KOR, respectively. A fourth group of tetrapeptides
(endomorphins) with yet-unknown precursors do not contain the pan-opioid motif, but bind to MOR with
high selectivity. Opioid peptides and receptors are expressed throughout the central and peripheral nervous
systems, in neuroendocrine tissues, and in immune cells (Roques, Fournie-Zaluski, & Wurm, 2012;
Schumacher et al., 2015; Stein & Machelska, 2011).

Opioid Receptor Signaling


ORs (and other GPCRs) have orthosteric and allosteric binding sites. The former are de ned as the sites for
endogenous opioid peptides and standard exogenous ligands, and the latter are separate sites for
endogenous or exogenous modulators (Livingston & Traynor, 2017; Wacker, Stevens, & Roth, 2017).
p. 737 Orthosteric ligand binding triggers intracellular coupling of the receptor to Gi/o and to other proteins
(e.g., arrestins). Receptor-induced Giα protein activation by guanosine triphosphate (GTP) evokes the
dissociation of the heterotrimeric G protein–complex into α, β, and γ subunits. Giα induces inhibition of
adenylyl cyclase and cyclic AMP (cAMP) production, while β and γ subunits interact with ion channels at the
+
plasma membrane (Williams et al., 2013). This leads to opening of G protein–coupled inwardly rectifying K
+
(GIRK) channels, as well as to the inhibition of Na channels, acid-sensing ion channels (ASICs), glutamate
++
receptor currents, and Ca in ux. Together, these events attenuate neuron excitability and the release of
pro-nociceptive neuropeptides, and thereby decrease the transmission of nociceptive stimuli and
(ultimately) pain perception (Stein, 2016). Other opioid-regulated pathways include phospholipase C and
mitogen-activated protein kinase (Pradhan, Smith, Kie er, & Evans, 2012; Stein, 2016; Williams et al.,
2013).

Upon activation, ORs can be phosphorylated by di erent kinases, thereby promoting β–arrestin
recruitment, receptor desensitization, and internalization, followed either by dephosphorylation and
recycling to the cell surface, or by degradation in lysosomes. Di erent ligands at the same receptor may
trigger distinct receptor conformations that can result in signaling through distinct intracellular pathways.
This has been termed “biased signaling” or “functional selectivity.” Experimental studies have suggested
that such preferential activation of G-proteins vs. β-arrestin may entail reduced adverse e ects, and that
this bias is ligand-speci c. For example, tolerance to morphine’s anti-nociceptive e ects and physical
dependence were decreased in beta-arrestin-2 knock-out (KO) mice, while responses to methadone,
fentanyl, or oxycodone were unchanged (Raehal, Schmid, Groer, & Bohn, 2011). The resolution of MOR,
DOR, and KOR crystal structures (Granier et al., 2012; Manglik et al., 2012; Wu et al., 2012) led to further
elucidation of ligand-induced conformational changes (Sounier et al., 2015) and to structure-based
screening of possible anti-nociceptive ligands (Manglik et al., 2016).

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


A novel example of “biased signaling” was demonstrated recently. In the acidotic environment of peripheral
injured tissue (Table 25.1), opioid receptor–ligand interactions apparently exhibit di erent dynamics
compared to non-injured tissues (Del Vecchio, Spahn, & Stein, 2017; Spahn et al., 2017). Computer
simulations indicated that opioid ligands assume a more stable binding position at low pH than at
physiological pH, suggesting that agonists have an enhanced potential to activate the receptor under acidic
(in amed) conditions. Based on these in silico studies, a prototype ligand (NFEPP) was designed that
exhibited preferential cAMP inhibition, G-protein dissociation, and anti-nociceptive e cacy in injured
tissue (at low pH), while typical adverse e ects (respiratory depression, reward, sedation, motor
disturbance, constipation) elicited in non-injured environments (at normal pH in brain or intestinal wall)
were absent, consistent with the notion that “normal” ORs were not activated (Figure 25.3) (Spahn et al.,
2017). These ndings were attributed both to acidosis-induced conformational alterations of peripheral
p. 738 ORs, and to the low acid dissociation constant (pKa) of NFEPP (Spahn et al., 2017).
Figure 25.3.

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


A novel concept to selectively activate opioid receptors in injured peripheral tissue without side e ects. NFEPP (purple circles)
was designed to act only at low pH in an inflamed environment (red) surrounding the peripheral nerve endings (light gray), by
decreased pKa and increased protonation (light blue triangles), a prerequisite for binding and activation of opioid receptors. The
protonated ligand binds and activates opioid receptors (dark gray); the unprotonated ligand (in healthy brain and
gastrointestinal wall) does not. NFEPP: (±)-N-(3-fluoro-1-phenethylpiperidine-4-yl)-N-phenylpropionamide (gray circle insert).

Reprinted with permission from G. Del Vecchio, V. Spahn, & C. Stein, Novel opioid analgesics and side e ects, ACS Chemical
Neuroscience, 8:1638–1640, © 2017, American Chemical Society.

p. 739 Tolerance and Opioid-Induced Hyperalgesia (OIH)


“Tolerance” describes the phenomenon that the magnitude of a given e ect decreases with repeated
administration of the same agonist dose, or that increasing doses are needed to produce the same e ect.
MOR agonists induce potent analgesia, but also side e ects, including nausea, constipation, respiratory
depression, and somnolence. All these e ects can be subject to tolerance development, albeit to di erent
degrees (Collett, 1998; McNicol, 2008; Rozen & Grass, 2005; Williams et al., 2013). Experimental evidence
suggests that some agonists may induce hyperalgesia (e.g., OIH) in animal models. Studies in mice with
MOR gene inactivation have shown that these diverse e ects are mediated by activating MOR (Gaveriaux-
Ru , 2013). Some studies suggest that OIH may contribute to analgesic tolerance, although tolerance and
OIH can be dissociated (Ferrini et al., 2013; Koblish et al., 2017).
Analgesic Tolerance
Pharmacodynamic tolerance to anti-nociceptive e ects is readily inducible in animal models but has not
been unequivocally demonstrated in controlled clinical studies. Following the activation of ORs, uncoupling
of G proteins, phosphorylation, beta-arrestin recruitment, and receptor endocytosis led to receptor
desensitization in vitro (Cahill, Walwyn, Taylor, Pradhan, & Evans, 2016; Williams et al., 2013). Several
approaches have been used to reduce tolerance development (Bruchas & Roth, 2016; Law et al., 2013; Olson,
Lei, Keresztes, LaVigne, & Streicher, 2017; Yaksh, Woller, Ramachandran, & Sorkin, 2015). These include
bivalent or multivalent ligands, biased agonists, and the design of novel peripherally acting opioid agonists
(see in Opioid Agonists and Analgesia).

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Tolerance development may be diminished in chronic pain (Pradhan et al., 2012). In a model of persistent
paw in ammation in rats, tolerance to morphine anti-nociception was evoked by the deletion of opioid
peptides in immune cells, indicating that endogenous OR activation promotes receptor endocytosis and
thereby counteracts tolerance (Zollner et al., 2008). Clinical studies were consistent with this notion (Stein
et al., 1996). The particular role of peripheral ORs in morphine tolerance was investigated by genetic
approaches in mice. The conditional KO (cKO) of MOR in nociceptive neurons expressing the sodium
channel Nav1.8 did not modify tolerance to morphine anti-nociception (Weibel et al., 2013). In comparison,
MOR deletion in neurons expressing the transient receptor potential vanilloid 1 receptor (TRPV1) abolished
anti-nociceptive tolerance (Corder et al., 2017). In rodent models of in ammatory and neuropathic pain,
morphine tolerance was prevented by administration of the peripherally restricted MOR antagonist
methylnaltrexone (Corder et al., 2017). To arrive at de nitive conclusions, further experiments in cKO mice
with persistent pain are required, and TRPV1 expression in brain needs to be considered (Madasu, Roche, &
Finn, 2015; Martins, Tavares, & Morgado, 2014).

p. 740 Opioid-Induced Hyperalgesia


Many animal studies have produced evidence for OIH. The putative underlying mechanisms are summarized
in recent reviews (Rivat & Ballantyne, 2016; Roeckel, Le Coz, Gaveriaux-Ru , & Simonin, 2016). Commonly
involved pathways comprise peripheral and central sensitization, descending facilitation, and
neuroimmune activation. There are more reports on mechanisms at the spinal level, but peripheral sites and
brain structures have also been investigated. At the cellular level, neurons, microglia, and astrocytes
participate (Ferrini et al., 2013). Mesenchymal stem cells transplanted into rats or mice were shown to
reverse tolerance and OIH (Hua et al., 2016). Toll-like receptors (Hutchinson et al., 2011) and MOR have been
shown to be involved in OIH (Corder et al., 2017; Roeckel et al., 2017). Other molecules comprise the TRP
family, the NMDA-glutamate system, substance P, ATP, K+/Cl– transporters, lipids, chemokines,
cytokines, serotonin, brain-derived neurotrophic factor (BDNF), and anti-opioid peptides (Elhabazi et al.,
2017; Rivat & Ballantyne, 2016; Roeckel et al., 2016).

Allosteric Modulators
Several allosteric modulators of ORs were investigated and shown to in uence the a nity and/or e cacy of
orthosteric ligands in vitro. However, in vivo con rmation is lacking so far (Livingston & Traynor, 2017). It
is an interesting concept that positive allosteric modulators may enhance the activity of endogenous opioid
peptides that are elevated during stress and pain. This activity would be con ned to ORs that are exposed to
released endogenous opioids and thereby avoid side e ects (similar to the concept of enkephalinase
inhibitors) (Livingston & Traynor, 2017; Roques et al., 2012).
Opioid Agonists and Analgesia
Many OR agonists have been developed since the identi cation of morphine as the active ingredient of
opium and the discovery of ORs (Brownstein, 1993). The opioids in clinical use are mostly MOR agonists and
have been reviewed in Drewes et al. (2013). They are e ective analgesics, although the variability of
responses among patients often requires a personalized approach (Drewes et al., 2013). ORs are expressed at
all levels of the neuraxis. Therefore, opioid agonists can inhibit pain following peripheral, spinal, cerebral,
and systemic administration.

Common examples of MOR, DOR, and KOR agonists and antagonists are shown in Table 25.2. More

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


extensive reviews on MOR (Drewes et al., 2013; Madariaga-Mazon et al., 2017), DOR (Peppin & Ra a, 2015;
Pradhan, Befort, Nozaki, Gaveriaux-Ru , & Kie er, 2011; Spahn & Stein, 2017), and KOR (Ranjan, Pandey, &
Shukla, 2017; Zheng et al., 2017) are available. New developments include the discovery and characterization
of bi-/multi-functional agonists, biased ligands, peripherally restricted agonists, and inhibitors of
endogenous opioid peptide degradation.

Table 25.2 Opioid Receptors and Ligands

Receptor Agonists Antagonists

MOR morphine, fentanyl, remifentanil, methadone, oxycodone, DAMGO, CTOP, CTAP, alvimopan
endomorphins, enkephalins, β-endorphin, TRV130, PZM21 naloxone (-methiodide)

DOR SNC80, enkephalins, β-endorphin, deltorphin, JNJ-20788560, ARM390, UPF-512, naltrindole naloxone (-
ADL5747, ADL5859, KNT-127 methiodide)

KOR U-50488, U69593, dynorphin nalfurafine, asimadoline norbinaltorphimine


naloxone (-methiodide)

p. 741 Bifunctional and Multifunctional Ligands


Bi-/multi-functional agonists were designed with the goal of reducing side e ects (Gunther et al., 2017;
Yekkirala et al., 2017). This strategy is based on evidence that OR form heteromers (Gomes et al., 2016;
Massotte, 2015) or interact with other GPCRs (Hubner et al., 2016; Olson et al., 2017; Yaksh et al., 2015). A
large number of such ligands have been investigated (Gunther et al., 2017; Olson et al., 2017; Yekkirala et al.,
2017) (Figure 25.4), and some were also evaluated for tolerance development in preclinical models. Less
tolerance was described for the MOR-agonist/DOR-agonist LP1; MOR-agonist/DOR-antagonists MDAN-21,
VRP26, SoRI22138, UMB425, and mitragynine/corynantheidine; MOR-agonist/CCK-antagonists RSA 504
and RSA 601; MOR-agonist/CCR5-antagonist MCC22; OR-agonist/NK1-antagonists TY027, RCCHM3, and
RCCHM6; MOR-agonist/mGluR5-antagonist MMG22; and the MOR-agonist/noradrenaline reuptake
blockers tapentadol and dezocine (Baron et al., 2017; Wang, Mao, Li, Gong, & Zhang, 2017).
Figure 25.4.

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Bi- and multi-valent opioid agonists. * CB2, cannabinoid receptor-2; CCK, cholecystokinin; DOR, delta opioid receptor; GPCR, G
protein-coupled receptor; KOR, kappa opioid receptor; mGlur5, metabotropic glutamate receptor-5; MOR, mu opioid receptor;
NK1, Neurokinin 1 receptor; NRI, noradrenalin reuptake inhibitor; OFQ, OrphaninFQ/nociceptin receptor.

MOR Agonists – GPCR Agonists: Compounds that activate two or more ORs include 3-[(2R,6R,11R)-8-
hydroxy-6,11-dimethyl-1,4,5,6-tetrahydro-2,6-methano-3-benzazocin-3(2H)-yl]-N-
phenylpropanamide, LP1 (Turnaturi, Arico, Ronsisvalle, Parenti, & Pasquinucci, 2016), morphine-6-O-
sulfate (Yadlapalli et al., 2016), biphalins (Mollica et al., 2014), N-Naphthoyl-betanaltrexamine (NNTA) and
N-2´-Indolylnaltrexamine 3 (INTA (Le Naour et al., 2014), DPI-125 (Yi et al., 2017), cyclotetrapeptide (De
Marco et al., 2016), dihydroetorphine (Gunther et al., 2017), BU08028, and SR16435 (Ding et al., 2016;
Gunther et al., 2017). The combination of MOR and cannabinoid agonists resulted in synergistic anti-
nociception (Grenald et al., 2017). In models of neuropathic and diabetic pain, the OR/OFQ agonist
cebranopadol prevented hyperalgesia (Tzschentke, Linz, Frosch, & Christoph, 2017), and a rst clinical
study showed its analgesic activity in patients (Christoph, Eerdekens, Kok, Volkers, & Freynhagen, 2017).

MOR Agonists -GPCR Antagonists: Based on previous literature showing reduced tolerance to morphine
anti-nociception by blocking DOR, several bivalent MOR-agonist/DOR-antagonists were investigated
(Fujita, Gomes, & Devi, 2015; Gendron, Cahill, von Zastrow, Schiller, & Pineyro, 2016; Hubner et al., 2016;
p. 742 Olson et al., 2017; Yaksh et al., 2015; Yekkirala et al., 2017). They comprise the DIPP- and PICPsi families,
MDAN-21, SoRI22138, UMB425 and UMB246 (Healy et al., 2017), VRP26 (Anand, Boyer, Mosberg, &
Jutkiewicz, 2016) and mitragynine/corynantheidine (Varadi et al., 2016) and others (Figure 25.4). These also
include molecules that activate MOR and are antagonists at sigma-1 (Prezzavento et al., 2017),
Nociceptin/OrphaninFQ (NOP/OFQ) (Lagard et al., 2017), cannabinoid, CCR5 and CXCR4 (Melik
Parsadaniantz, Rivat, Rostene, & Reaux-Le Goazigo, 2015), neurokinin-1 (Starnowska et al., 2017),
glutamate mGluR5, and cholecystokinin receptors (Figure 25.4).

MOR Agonists – Noradrenalin Reuptake Inhibitors: Tramadol produces analgesia by activating MOR and
blocking noradrenaline/serotonin reuptake, but it requires metabolism by CYP2D6 and has been shown to
cause confusion in the clinical setting. Tapentadol analgesia requires MOR activation and norepinephrine
reuptake inhibition, but no metabolic activation (Langford, Knaggs, Farquhar-Smith, & Dickenson, 2016).
Dezocine (a popular drug in China) produces anti-nociception in rats by activating MOR and blocking
norepinephrine reuptake (Wang et al., 2017).
Biased Agonists
The knowledge gain on biased signaling and OR structures has led to extensive screening e orts for
discovery of new ligands and has broadened the general view on GPCR function (Bruchas & Roth, 2016;
p. 743 Wacker et al., 2017). New MOR, DOR, or KOR ligands were shown to preferentially activate G-proteins
over beta-arrestins in vitro and to evoke anti-nociception in preclinical models in vivo (Koblish et al., 2017;
Madariaga-Mazon et al., 2017; Violin, Crombie, Soergel, & Lark, 2014). The Gi-biased MOR agonists TRV130
and PZM21 exerted low beta-arrestin recruitment, anti-nociception, and reduced adverse e ects in rodent
models (Altari et al., 2017; DeWire et al., 2013; Kie er, 2016; A. Manglik et al., 2016). Repeated TRV130
application did not induce anti-nociceptive tolerance in animals (Altari et al., 2017; DeWire et al., 2013; A.

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Manglik et al., 2016). In healthy human volunteers, TRV130 produced inhibition of experimental pain, less
respiratory depression and vomiting, but more nausea, dizziness, somnolence, and headache than
morphine (Soergel et al., 2014). In further animal (Altari et al., 2017) and human clinical studies (Viscusi et
al., 2016), TRV130 produced adverse e ects similar to those of conventional opioids; that is, nausea,
dizziness, vomiting, and constipation. In mice, another biased MOR agonist (TRV0109101) induced anti-
nociception, tolerance, and constipation, but no OIH, and it reversed morphine- and fentanyl-induced OIH
but not fentanyl-induced tolerance (Koblish et al., 2017).

Biased agonism was also shown for KOR, both in vitro and in preclinical models. The KOR agonists U50488
and salvinorin di ered in downstream kinase activation. U50488 was shown to regulate extracellular-
regulated kinase, while salvinorin elicited c-Jun kinase and induced homologous desensitization (Jamshidi
et al., 2015). A few new G-protein-biased KOR ligands produced anti-nociception and fewer unwanted KOR
e ects (Ranjan et al., 2017). Ibogaine potentiated morphine anti-nociception, prevented morphine
tolerance and exhibited anti-addictive e ects (Maillet et al., 2015). RB-64 induced anti-nociception and
aversion but no sedation (White et al., 2015). Triazole 1.1 and HS666 elicited anti-nociception with reduced
sedation, without dysphoria or decrease of dopamine tone in the nucleus accumbens (Brust et al., 2016;
Spetea et al., 2017). These preclinical studies suggest the potential of G-protein-biased KOR agonists.

There is also ample evidence for biased agonism at DOR (Cahill et al., 2016; Gendron et al., 2016; Vicente-
Sanchez & Pradhan, 2018). This has been reviewed with scrutiny and emphasis on ligand-speci c signaling
and receptor regulation (Gendron et al., 2016). In the context of anti-nociception, DOR internalization,
desensitization, resensitization, as well as coupling to beta-arrestins and kinases were studied (Cahill et al.,
2016). Despite disparities in receptor signaling and e ects on intracellular tra cking, practically all DOR
agonists exhibited anti-nociceptive tolerance (Gendron et al., 2016; Nozaki et al., 2014), with the exception
of JNJ-20788560. Contrasting data have been reported for ARM390 (Codd et al., 2009; Pradhan et al., 2016).
Peripheral Opioid Receptors and Agonists
Over 25 years ago, evidence began to emerge that signi cant anti-nociceptive e ects are mediated by ORs
localized on peripheral sensory neurons, and that opioid agonists elicit stronger analgesic e ects in
in amed than nonin amed tissue of animals and humans (Stein, 1993, 1995; Stein et al., 1991; Stein et al.,
1990). These observations stimulated extensive research into the underlying mechanisms. It was found that
p. 744 peripheral tissue in ammation induced upregulation of ORs and their mRNAs in DRG neurons, which
was dependent on neuronal electrical activity and on local cytokine production (Figure 25.1) (Stein, 2016;
Stein & Machelska, 2011). Studies in those models also showed that the peripherally directed axonal
transport of ORs in DRG neurons was increased (Hassan, Ableitner, Stein, & Herz, 1993) and that the

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


perineural barrier was disrupted, thus facilitating access of opioid agonists to their receptors (Antonijevic,
Mousa, Schäfer, & Stein, 1995). These events were ascribed to the in uence of various in ammatory
mediators such as bradykinin, nerve growth factor, and prostaglandins (Stein, 2016; Stein & Machelska,
2011). Furthermore, it was shown that G-protein coupling of ORs was augmented (Zöllner et al., 2003), and
that low pH (as in in ammation, see Table 25.1) increased opioid agonist e cacy in vitro (Rasenick &
Childers, 1989; Selley, Breivogel, & Childers, 1993). Recordings from sensory nerve bers supplying injured
tissue revealed opioid inhibition of spontaneous and stimulus-evoked action potentials (reviewed in Stein,
2016). Neuropathy is another condition in uencing opioid receptor expression in peripheral sensory
neurons (Machelska, 2011; Stein & Machelska, 2011). For example, upregulation of ORs and accumulation of
opioid peptide-producing immune cells was detected at the site of nerve injury, accompanied by enhanced
anti-nociceptive activity of opioid agonists (Celik et al., 2016; Stein, 2016). Thus, the expression, axonal
transport, signaling, and accessibility of ORs on DRG neurons are augmented, suggesting that tissue or
nerve injury are prerequisites to “unmasking” peripheral opioid e ects (Stein, 1993, 2016). Opioid peptides
and receptors are also expressed by immune cells (Sharp, 2006; Stein & Machelska, 2011), and there is
evidence that they contribute to analgesia (Celik et al., 2016).

Peripherally acting agonists were developed with the idea that OR activation at the site of pain generation
will trigger analgesia without the adverse e ects mediated by ORs in the CNS (Stein, 2013a). This concept
has been investigated extensively by pharmacological approaches and by using cKO mice (reviewed in Stein,
1993, 2016; Stein & Machelska, 2011). For example, the absence of mu ORs in peripheral nociceptive neurons
did not change morphine anti-nociception in models of acute pain (Corder et al., 2017; Weibel et al., 2013),
but decreased morphine and fentanyl antihyperalgesia in a model of persistent paw in ammation,
indicating that peripheral MOR signi cantly contribute to analgesia produced by systemically administered
opioids (Weibel et al., 2013). The absence of DOR in Nav1.8 neurons aggravated both in ammatory and
neuropathic hypersensitivity, and greatly lowered DOR agonist-induced anti-nociception in models of
chronic pain (Gaveriaux-Ru & Kie er, 2011; Nozaki et al., 2012).

Recently, a novel agonist (NFEPP) was designed based on computational simulations of ligand-receptor
interactions in an in amed (acidic) environment. This agonist selectively activated MOR at acidic pH and
induced peripherally mediated, injury-speci c anti-nociception without a ecting locomotor activity,
reward, bowel movements, or respiration (Spahn et al., 2017). Another novel peripherally acting analgesic is
morphine covalently attached to hyperbranched polyglycerol (PG-M). This conjugate was shown to release
morphine selectively in injured tissue. PG-M produced peripherally mediated analgesia in a model of
p. 745 in ammatory pain without eliciting sedation or constipation (Gonzalez-Rodriguez et al., 2017). Both
NFEPP and PG-M produced equie ective anti-nociception to conventional opioid agonists.
Endogenous Analgesia: Central Mechanisms
Spinal cord interneurons produce opioids that, together with noradrenergic and serotonergic pathways,
contribute to the descending pain inhibition from the brainstem. An endogenous opioid analgesic tone was
revealed in mutant mice lacking individual components of the opioid system. DOR-KO mice show
aggravated neuropathic, in ammatory, and visceral hypersensitivity (Gaveriaux-Ru , Karchewski, Hever,
Matifas, & Kie er, 2008; Nadal, Banos, Kie er, & Maldonado, 2006; Reiss et al., 2017). In ammatory
hypersensitivity was comparable or longer lasting in MOR-KO (Gaveriaux-Ru et al., 2008; Mansikka,
Zhou, Donovan, Pertovaara, & Raja, 2002; Walwyn et al., 2016). An endogenous tone at MOR was shown in
neuropathic mechanical allodynia, but not in thermal hyperalgesia (Mansikka et al., 2004). In addition,

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


MOR-KO animals showed augmented gray matter volume of periaqueductal and altered connections of
pain-aversion nodes (Mechling et al., 2016; Sasaki et al., 2015). Mice lacking KOR showed augmented
sensitivity to visceral chemical stimulation (Simonin et al., 1998), neuropathic pain (Xu et al., 2004),
unchanged response to colorectal distension (Larsson, Bayati, Lindstrom, & Larsson, 2008), and augmented
in ammatory hypersensitivity (Gaveriaux-Ru et al., 2008; Schepers, Mahoney, Gehrke, & Shippenberg,
2008). An additive contribution to the anti-nociceptive tone at each receptor was found in triple
MOR/DOR/KOR-KO animals (Gaveriaux-Ru , 2013). Beta-endorphin-KO and preproenkephalin-KO mice
showed normal acute nociception and in ammatory pain (Kie er & Gaveriaux-Ru , 2002; Labuz, Celik,
Zimmer, & Machelska, 2016; Walwyn et al., 2016). Prodynorphin KO animals display normal in ammatory
pain (Walwyn et al., 2016), recover more rapidly from neuropathic hypersensivity (Kie er & Gaveriaux-
Ru , 2002; Xu et al., 2004), and are slightly less sensitive after nerve injury (Labuz et al., 2016). Endogenous
opioids also participate in bromyalgia. In a mouse bromyalgia model, electroacupuncture-induced
analgesia was abolished by the antagonist naloxone (Yen, Hsieh, Hsu, & Lin, 2017). It was also found that
MOR availability was reduced in brain regions of bromyalgia patients (Schrepf et al., 2016), and that
endogenous opioids contribute to placebo analgesia (Carlino, Frisaldi, & Benedetti, 2014; Holmes, Tiwari, &
Kennedy, 2016), to the e ects of transcranial magnetic stimulation (Lamusuo et al., 2017), and to congenital
insensitivity to pain caused by a Nav1.7-null mutation (Minett et al., 2015).

Peripheral Mechanisms
Opioid peptide-containing immune cells extravasate and accumulate in peripheral injured tissues
(Baddack-Werncke et al., 2017; Cayla et al., 2012; Celik et al., 2016; Labuz et al., 2009; Mousa, Cheppudira, et
al., 2007; Stein, 2013b; Stein & Machelska, 2011). These cells upregulate the gene expression of opioid
peptide precursors and the enzymatic machinery for their processing into functionally active peptides
p. 746 (Busch-Dienstfertig, Labuz, Wolfram, Vogel, & Stein, 2012; Mousa, Shakibaei, Sitte, Schäfer, & Stein,
2004; Sitte et al., 2007). In response to stress, catecholamines, corticotropin releasing factor, cytokines,
chemokines, opioid ligands or bacteria, leukocytes secrete opioid peptides, which then activate peripheral
ORs and produce analgesia by inhibiting the excitability of nociceptors, the release of excitatory
neuropeptides, or both (Celik et al., 2016; Rittner et al., 2009; Stein & Machelska, 2011).

Further studies showed that endogenous opioid anti-nociception was dampened by naloxone or by T-
lymphocyte depletion in a mouse model of in ammatory pain. Proenkephalin was expressed in T-
lymphocytes and dendritic cells (Boue, Blanpied, Brousset, Vergnolle, & Dietrich, 2011) and, more precisely,
in Th1 and Th2 CD4 cells (Boue et al., 2012). In a similar study in rats, proopiomelanocortin transcripts and
beta-endorphin were expressed by B-lymphocytes in lymph nodes (Maddila, Busch-Dienstfertig, & Stein,
2017). In rodent models of visceral pain, endogenous pain regulation was mediated by CD4 lymphocyte-
derived opioids (Boue et al., 2014; Valdez-Morales et al., 2013) and, more speci cally, by enkephalins (Basso
et al., 2017). There is also evidence for endogenous analgesia mediated by opioid released from CD8
lymphocytes in a mouse model of antigen-collagen-induced arthritis (Baddack-Werncke et al., 2017). Anti-
nociception elicited by corticotropin-releasing-factor administered onto damaged nerves of wild-type mice
was absent in mice lacking proenkephalin, prodynorphin, or beta-endorphin (Labuz et al., 2016). Polarized
M2 macrophages were shown to release high amounts of opioid peptides to induce anti-nociception in this
neuropathy model (Pannell et al., 2016). In response to exogenous opioids, opioid peptides produced by
immune cells at the nerve lesion site produce anti-nociception, as shown by studies in opioid peptide- and
OR-KO mice (Celik et al., 2016).

The clinical relevance of these mechanisms has been con rmed in studies demonstrating that patients with
knee joint in ammation express opioid peptides in immune cells and ORs on sensory nerve terminals within
synovial tissue (Mousa, Straub, Schäfer, & Stein, 2007; Stein, Hassan, Lehrberger, Gie ng, & Yassouridis,
1993). After knee surgery, pain and analgesic consumption were increased by blocking the interaction

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


between the endogenous opioids and their receptors with intraarticular naloxone or adrenergic antagonists
(Kager et al., 2011; Stein et al., 1993), and were diminished by stimulating opioid peptide secretion (Likar et
al., 2007) or by intraarticular morphine administration (Stein et al., 1991; Zeng et al., 2013).

Augmenting Endogenous Opioid Analgesia


Opioid peptides are susceptible to rapid extracellular degradation by aminopeptidase N and neutral
endopeptidase (“enkephalinases”). Preventing this degradation by inhibitors (in the CNS or in peripheral
tissues) has been shown to produce analgesic e ects in many animal models and in some human trials
(Bonnard et al., 2015; Roques et al., 2012; Schreiter et al., 2012). This strategy avoids unphysiologically high
concentrations of exogenous agonists at (ubiquitously distributed) receptors, and, thus, diminishes the risk
for development of receptor downregulation, tolerance, desensitization, o -site, or paradoxical excitatory
p. 747 e ects (Roques et al., 2012). The combination of the peptidase inhibitors bestatin and thiorphan
(Schreiter et al., 2012) and the dual enkephalinase inhibitor PL265 inhibited hyperalgesia in preclinical
models of in ammatory and neuropathic pain (Bonnard, Poras, Fournie-Zaluski, & Roques, 2016; Bonnard
et al., 2015; Poras, Bonnard, Fournie-Zaluski, & Roques, 2015). Other preclinical approaches used
administration of viruses or cells overexpressing opioid peptides (Goins, Cohen, & Glorioso, 2012; Guedon et
al., 2015). The human natural peptide opiorphin protects enkephalins from degradation. It produced anti-
nociception equipotent to morphine (Wisner et al., 2006), displayed no constipating e ects or abuse
liability (Popik, Kamysz, Kreczko, & Wrobel, 2010; Rougeot, Robert, Menz, Bisson, & Messaoudi, 2010), and
produced antidepressant e ects via DOR activation (Javelot, Messaoudi, Garnier, & Rougeot, 2010; Rougeot
et al., 2010). Opiorphin loading of liposomes improved its activity (Mennini et al., 2015). Recently, STR-324,
a stable analog of opiorphin, was found to produce anti-nociception in a model of incisional
hypersensitivity (Sitbon et al., 2016).
Species Di erences
Species di erences are an important consideration (Chan, McCarthy, Li, Palczewski, & Yuan, 2017; Imam,
Kuo, Ghassabian, & Smith, 2017; Pasternak & Pan, 2013; Stevens, 2015). Numerous studies on intra- and
inter-species di erences have demonstrated that even single amino acid variations in OR proteins can have
measurable e ects on the structure-activity of the receptor (Busch-Dienstfertig et al., 2013; Nockemann et
al., 2013; Pasternak & Pan, 2013; Stevens, 2015; Vardy et al., 2015). Extensive alternative splicing of the MOR
gene has been described in di erent species and strains, possibly enabling divergent anatomical
distributions, expression levels, oligomers, recycling, and intracellular signaling events (Pasternak & Pan,
2013). In addition, disparate (sometimes opposite) CNS or intestinal phenomena were detected in mice

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


versus rats or humans (e.g., G-protein activation, adenylyl cyclase, locomotion) (Fidecka, Malec, &
Langwinski, 1978; Huang, Chen, & Liu-Chen, 2015; Imam et al., 2017; Kruegel et al., 2016; Labuz, Mousa,
Schäfer, Stein, & Machelska, 2007; Noble & Cox, 1995, 1996; Sirohi, Aldrich, & Walker, 2016). For example,
chronic DOR activation led to OIH in rats but not in mice, whereas MOR-induced OIH occurred in all tested
species (Rowan et al., 2014). A recent study reported that tolerance to opioid-induced analgesia was
dependent on MOR expressed in DRG neurons in mice (Corder et al., 2017), contrary to ndings in rats and
+
humans (Stein et al., 1996; Zollner et al., 2008). Opioid agonists did not a ect K currents (Dembla et al.,
2017; Nockemann et al., 2013) or TRPV1 channels (Dembla et al., 2017; Quallo, Alkhatib, Gentry, Andersson,
& Bevan, 2017; Rowan et al., 2014) in mouse DRG neurons, but they did so in rat (Endres-Becker et al., 2007;
+
Nockemann et al., 2013; Rowan et al., 2014; Spahn et al., 2013). Studies on gene expression of K channels
suggested a greater similarity between humans and rats than between humans and mice (Nockemann et al.,
2013). As in other elds (e.g., immunology, oncology), all these ndings have to be taken into consideration
when preclinical data are interpreted to predict drug e ects in humans.

p. 748
Clinical Problems Associated with Opioid Use

In contrast to the experimental literature, there is a lack of carefully controlled clinical studies that
unequivocally demonstrate the development of pharmacodynamic tolerance in opioid analgesia (Galer, Lee,
Ma, Nagle, & Schlagheck, 2005; Schneider & Kirsh, 2010). Incomplete cross-tolerance between opioids may
explain clinical observations that switching drugs (“opioid rotation”) is occasionally useful in patients with
inadequate pain relief or intolerable side e ects (Ross, Riley, Quigley, & Welsh, 2006). Importantly,
pharmacokinetic (e.g., altered distribution or metabolism of the opioid) and learned tolerance (e.g.,
compensatory skills developed during mild intoxication), as well as increased nociceptive stimulation by
tumor growth, in ammation, or neuroma formation, are possible reasons for increased dose requirements
(Collett, 1998; Collin, Poulain, Gauvain-Piquard, Petit, & Pichard-Leandri, 1993).

There is an ongoing debate whether opioids paradoxically induce hyperalgesia (OIH) in humans. At high
doses, occasionally encountered in extreme cancer pain, allodynia has been observed and attributed to
neuroexcitatory e ects of opioid metabolites (Carullo, Fitz-James, & Delphin, 2015). Similar phenomena
have also been reported in patients with chronic non-malignant pain (e.g., migraine) (Belkin, Reinheimer,
Levy, & Johnson, 2017; Diener, Holle, Solbach, & Gaul, 2016; Gri n et al., 2016; Wasserman et al., 2015),
postoperative pain (Fletcher & Martinez, 2014), in healthy volunteers (Fishbain, Cole, Lewis, Gao, &
Rosomo , 2009; Mauermann et al., 2016; Ruscheweyh, Wilder-Smith, Drdla, Liu, & Sandkuhler, 2011), and
in opioid addicts (Compton, Charuvastra, & Ling, 2001). Upon closer scrutiny, it appears that most studies
have in fact shown withdrawal-induced hyperalgesia, a well-known phenomenon following the abrupt
cessation of opioids (Comelon et al., 2016; Fishbain et al., 2009; Spahn et al., 2013). With the possible
exception of headache (Diener et al., 2016), conclusive evidence is lacking that clinically signi cant OIH
occurs during the perioperative or chronic administration of usual opioid doses in patients (Angst, 2015;
Fishbain et al., 2009; Fletcher & Martinez, 2014; Schneider & Kirsh, 2010).
Dependence is not synonymous with tolerance. Physical dependence is de ned as a state of adaptation that
is manifested by a withdrawal syndrome elicited by abrupt cessation, rapid dose reduction, and/or
administration of an antagonist (Smith et al., 2013). All opioids produce clinically relevant physical
dependence, even when administered for only a relatively short period of time (Compton, Miotto, &
Elasho , 2004).

Addiction is a complex syndrome involving reward/euphoria, the urge to avoid withdrawal, craving,
uncontrolled/compulsive drug use despite harmful side e ects, and other drug-related aberrant behaviors
(e.g., altering prescriptions, manipulating healthcare providers, drug hoarding, or unsanctioned dose
escalation) (Smith et al., 2013). Currently, an estimated 2 million individuals in the United States have

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


opioid use disorder (addiction) associated with prescription opioids (Psaty & Merrill, 2017; Schuchat, Houry,
& Guy, 2017).

p. 749 Long-Term Opioid Use in Chronic Pain


In contrast to acute and cancer-related pain, the long-term use of opioids in chronic non-cancer (e.g.,
neuropathic, musculoskeletal) pain is not appropriate. So far, randomized controlled trials (RCTs) have only
been conducted for a maximum duration of three months. In meta-analyses, the reduction of pain scores
was clinically insigni cant, and epidemiological data suggest that quality of life or functional capacity are
not improved (Eriksen, Sjogren, Bruera, Ekholm, & Rasmussen, 2006; Reinecke et al., 2015). Adverse side
e ects (nausea, sedation, constipation, dizziness, etc.) and lack of analgesic e cacy led to the dropout of
high numbers of subjects, both in RCTs and in uncontrolled observational studies beyond three months
(Gustavsson et al., 2012; Noble et al., 2010; Reinecke et al., 2015; Szigethy, Knisely, & Drossman, 2017).
Psychosocial outcome parameters were rarely investigated and showed only modest improvement. Thus,
consistent with the multifactorial nature of chronic pain, it is highly unlikely that opioids alone can produce
an analgesic response if, for example, there is a major a ective component or if learned pain behavior is the
main problem (Fordyce, 1991; Stein, 1997). Notwithstanding, opioids are prescribed widely, and addiction,
overdoses, death rates, and misuse have reached epidemic proportions (Psaty & Merrill, 2017; Schuchat et
al., 2017; Szigethy et al., 2017). Thus, the use of opioids as a sole treatment modality in chronic non-
malignant pain is strongly discouraged. Instead, chronic pain requires a multidisciplinary approach
encompassing various pharmacological and non-pharmacological (psychological, physiotherapeutic)
treatment strategies (Schuchat et al., 2017; Stein, 2013a; Szigethy et al., 2017).

Conclusion

Basic research on pain and analgesia continues at a rapid pace, but translation of ndings into clinical
applications has been di cult (Richards & McMahon, 2013; Yekkirala et al., 2017). Not only therapeutic but
also diagnostic approaches (e.g., brain imaging, genetics) are being investigated, but these have only rarely
reached practical applicability in patients (Chidambaran et al., 2017; Davis et al., 2017; Lee et al., 2012; Matic
et al., 2017; Mogil et al., 2010; Walter et al., 2013). Many obstacles have been discussed (Berge, 2011; Galer et
al., 2005; Mogil et al., 2010; Richards & McMahon, 2013; Yekkirala et al., 2017). Notwithstanding, animal
studies are indispensable, continue to be improved, and have successfully predicted adverse side e ects of
drug candidates (Berge, 2011; Mogil et al., 2010).

Areas of intense research are signaling, tra cking, and processing of ORs in sensory neurons, particularly
the in uence of injury and the implications for tolerance development, e cacy, and potency of opioid
agonists. The recent urry of publications on GPCR structures enables novel approaches to elucidate biased
signaling and allosteric and oligomeric modulation of opioid receptor function. A eld that has not received
p. 750 much attention is the in uence of opioids on in ammation and wound healing, despite ample preclinical
data (Stein & Küchler, 2013). Similarly, clinical studies on the augmentation of endogenous opioid e ects
are needed (Livingston & Traynor, 2017; Roques et al., 2012; Schreiter et al., 2012).

The epidemic of opioid misuse illustrates the persistent problems resulting from non-selective activation of
ubiquitous ORs throughout central and peripheral compartments. Thus, the potential of peripheral actions
is increasingly recognized by researchers and clinicians (Baron et al., 2013; Piomelli & Sasso, 2014; Richards
& McMahon, 2013; Roques et al., 2012; Stein, 2016; Vadivelu, Mitra, & Hines, 2011; Valverde & Gunkel, 2005;
Yekkirala et al., 2017; Zeng et al., 2013). These endeavors should eventually lead to novel pain medications
with fewer side e ects. In the wider context of pain management, however, one must not overlook the
in uence of psycho-social factors and the important role of non-pharmacological therapeutic approaches

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


(Fordyce, 1992; Schuchat et al., 2017; Stein, 1997, 2013a). It is therefore important that theoretical,
experimental, and clinical researchers establish direct contacts and that such collaborations be nurtured by
academic institutions, funding bodies, and industry.

Acknowledgments

This work was supported by Bundesministerium für Bildung und Forschung (0316177B/C1, 01EC1403E,
01EC1403F) (CS), the Helmholtz Virtual Institute “Multifunctional Biomaterials for Medicine” (CS), and the
University of Strasbourg (CG-R). This work has also been funded by the European Union Seventh
Framework programme (FP7-Health-2013-Innovation) under grant agreements 1602919 (CG-R) and
602891 (CS and CG-R).
References

Alfaro, V., Rodenas, J., Pesquero, J., Mitjavila, M. T., Palacios, L., & Carbonell, T. (1996). Factors influencing the acid-base changes
in the air-pouch exudate following carrageenan induced inflammation in rats. Inflammation Research, 45(8), 405–411. Retrieved
from http://www.ncbi.nlm.nih.gov/pubmed/8872514
Google Scholar WorldCat

Altarifi, A. A., David, B., Muchhala, K. H., Blough, B. E., Akbarali, H., & Negus, S. S. (2017). E ects of acute and repeated treatment
with the biased mu opioid receptor agonist TRV130 (oliceridine) on measures of ant nociception, gastrointestinal function, and
abuse liability in rodents. Journal of Psychopharmacology, 31(6), 730–739. doi:10.1177/0269881116689257

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Google Scholar WorldCat

Anand, J. P., Boyer, B. T., Mosberg, H. I., & Jutkiewicz, E. M. (2016). The behavioral e ects of a mixed e icacy anti-nociceptive
peptide, VRP26, following chronic administration in mice. Psychopharmacology (Berlin), 233(13), 2479–2487.
doi:10.1007/s00213-016-4296-8
Google Scholar WorldCat

Andersson, S. E., Lexmuller, K., Johansson, A., & Ekstrom, G. M. (1999). Tissue and intracellular pH in normal periarticular so
tissue and during di erent phases of antigen induced arthritis in the rat. Journal of Rheumatology, 26(9), 2018–2024. Retrieved
from http://www.ncbi.nlm.nih.gov/pubmed/10493685
Google Scholar WorldCat

Angst, M. S. (2015). Intraoperative Use of Remifentanil for TIVA: Postoperative Pain, Acute Tolerance, and Opioid-Induced
Hyperalgesia. Journal of Cardiothoracic & Vascular Anesthesia, 29(Suppl 1), S16–S22. doi:10.1053/j.jvca.2015.01.026
Google Scholar WorldCat

p. 751 Antonijevic, I., Mousa, S. A., Schäfer, M., & Stein, C. (1995). Perineurial defect and peripheral opioid analgesia in inflammation.
The Journal of Neuroscience, 15(1), 165–172.
Google Scholar WorldCat

Ashby, B. S. (1966). pH studies in human malignant tumours. Lancet, 2(7458), 312–315. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/4161494
Google Scholar WorldCat

Baddack-Werncke, U., Busch-Dienstfertig, M., Gonzalez-Rodriguez, S., Maddila, S. C., Grobe, J., Lipp, M., … Muller, G. (2017).
Cytotoxic T cells modulate inflammation and endogenous opioid analgesia in chronic arthritis. Journal of Neuroinflammation,
14(1), 30. doi:10.1186/s12974-017-0804-y
Google Scholar WorldCat

Baron, R. (2006). Mechanisms of disease: Neuropathic pain—a clinical perspective. Nature Clinical Practice. Neurology, 2(2), 95–
106. doi:10.1038/ncpneuro0113
Google Scholar WorldCat

Baron, R., Eberhart, L., Kern, K. U., Regner, S., Rolke, R., Simanski, C., & Tolle, T. (2017). Tapentadol prolonged release for chronic
pain: A review of clinical trials and 5 years of routine clinical practice data. Pain Practice: The O icial Journal of the World Institute
of Pain, 17(5), 678–700. doi:10.1111/papr.12515
Google Scholar WorldCat

Baron, R., Hans, G., & Dickenson, A. H. (2013). Peripheral input and its importance for central sensitization. Annals of Neurology,
74(5), 630–636. doi:10.1002/ana.24017
Google Scholar WorldCat

Basbaum, A. I., Bautista, D. M., Scherrer, G., & Julius, D. (2009). Cellular and molecular mechanisms of pain. Cell, 139(2), 267–284.
doi:10.1016/j.cell.2009.09.028
Basso, L., Garnier, L., Bessac, A., Boue, J., Blanpied, C., Cenac, N., … Dietrich, G. (2017). T-lymphocyte-derived enkephalins
reduce Th1/Th17 colitis and associated pain in mice. Journal of Gastroenterology, 53, 215–226. doi:10.1007/s00535-017-1341-2
Google Scholar WorldCat

Belkin, M., Reinheimer, H. S., Levy, J., & Johnson, B. (2017). Ameliorative response to detoxification, psychotherapy, and medical
management in patients maintained on opioids for pain. American Journal of Addiction, 26(7), 738–743. doi:10.1111/ajad.12605
Google Scholar WorldCat

Berge, O. G. (2011). Predictive validity of behavioural animal models for chronic pain. British Journal Of Pharmacology, 164(4),

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


1195–1206. doi:10.1111/j.1476-5381.2011.01300.x
Google Scholar WorldCat

Bonnard, E., Poras, H., Fournie-Zaluski, M. C., & Roques, B. P. (2016). Preventive and alleviative e ects of the dual enkephalinase
inhibitor (Denki) PL265 in a murine model of neuropathic pain. European Journal of Pharmacology, 788, 176–182.
doi:10.1016/j.ejphar.2016.05.041
Google Scholar WorldCat

Bonnard, E., Poras, H., Nadal, X., Maldonado, R., Fournie-Zaluski, M. C., & Roques, B. P. (2015). Long-lasting oral analgesic e ects
of N-protected aminophosphinic dual ENKephalinase inhibitors (DENKIs) in peripherally controlled pain. Pharmacology
Research & Perspectives, 3(2), e00116. doi:10.1002/prp2.116
Google Scholar WorldCat

Boue, J., Basso, L., Cenac, N., Blanpied, C., Rolli-Derkinderen, M., Neunlist, M., … Dietrich, G. (2014). Endogenous regulation of
visceral pain via production of opioids by colitogenic CD4(+) T cells in mice. Gastroenterology, 146(1), 166–175.
doi:10.1053/j.gastro.2013.09.020
Google Scholar WorldCat

Boue, J., Blanpied, C., Brousset, P., Vergnolle, N., & Dietrich, G. (2011). Endogenous opioid-mediated analgesia is dependent on
adaptive T cell response in mice. Journal of Immunology, 186(9), 5078–5084. doi:10.4049/jimmunol.1003335
Google Scholar WorldCat

Boue, J., Blanpied, C., Djata-Cabral, M., Pelletier, L., Vergnolle, N., & Dietrich, G. (2012). Immune conditions associated with CD4+
T e ector-induced opioid release and analgesia. Pain, 153(2), 485–493. doi:10.1016/j.pain.2011.11.013
Google Scholar WorldCat

Brownstein, M. J. (1993). A brief history of opiates, opioid peptides, and opioid receptors. Proceedings of the National Academy of
Sciences USA, 90(12), 5391–5393. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/8390660
Google Scholar WorldCat

Bruchas, M. R., & Roth, B. L. (2016). New technologies for elucidating opioid receptor function. Trends in Pharmacological
Sciences, 37(4), 279–289. doi:10.1016/j.tips.2016.01.001
Google Scholar WorldCat

p. 752 Bruehl, S., Apkarian, A. V., Ballantyne, J. C., Berger, A., Borsook, D., Chen, W. G., … Lin, Y. (2013). Personalized medicine and
opioid analgesic prescribing for chronic pain: Opportunities and challenges. Journal of Pain, 14(2), 103–113.
doi:10.1016/j.jpain.2012.10.016
Google Scholar WorldCat

Brust, T. F., Morgenweck, J., Kim, S. A., Rose, J. H., Locke, J. L., Schmid, C. L., … Bohn, L. M. (2016). Biased agonists of the kappa
opioid receptor suppress pain and itch without causing sedation or dysphoria. Science Signaling, 9(456), ra117.
doi:10.1126/scisignal.aai8441
Google Scholar WorldCat
Busch-Dienstfertig, M., Labuz, D., Wolfram, T., Vogel, N. N., & Stein, C. (2012). JAK-STAT1/3-induced expression of signal
sequence-encoding proopiomelanocortin mRNA in lymphocytes reduces inflammatory pain in rats. Molecular Pain, 8, 83.
doi:10.1186/1744-8069-8-83
Google Scholar WorldCat

Busch-Dienstfertig, M., Roth, C. A., & Stein, C. (2013). Functional characteristics of the naked mole rat mu-opioid receptor. PLoS
ONE, 8(11), e79121. doi:10.1371/journal.pone.0079121
Google Scholar WorldCat

Cahill, C. M., Walwyn, W., Taylor, A. M., Pradhan, A. A., & Evans, C. J. (2016). Allostatic mechanisms of opioid tolerance beyond
desensitization and downregulation. Trends in Pharmacological Sciences, 37(11), 963–976. doi:10.1016/j.tips.2016.08.002

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Google Scholar WorldCat

Carlino, E., Frisaldi, E., & Benedetti, F. (2014). Pain and the context. Nature Reviews Rheumatology, 10(6), 348–355.
doi:10.1038/nrrheum.2014.17
Google Scholar WorldCat

Carlino, E., Pollo, A., & Benedetti, F. (2011). Placebo analgesia and beyond: A melting pot of concepts and ideas for neuroscience.
Current Opinion in Anesthesiology, 24(5), 540–544. doi:10.1097/ACO.0b013e328349d0c2
Google Scholar WorldCat

Carullo, V., Fitz-James, I., & Delphin, E. (2015). Opioid-induced hyperalgesia: A diagnostic dilemma. Journal of Pain & Palliative
Care Pharmacotherapy, 29(4), 378–384. doi:10.3109/15360288.2015.1082006
Google Scholar WorldCat

Cayla, C., Labuz, D., Machelska, H., Bader, M., Schäfer, M., & Stein, C. (2012). Impaired nociception and peripheral opioid anti-
nociception in mice lacking both kinin B1 and B2 receptors. Anesthesiology, 116(2), 448–457.
doi:10.1097/ALN.0b013e318242b2ea
Google Scholar WorldCat

Celik, M. O., Labuz, D., Henning, K., Busch-Dienstfertig, M., Gaveriaux-Ru , C., Kie er, B. L., … Machelska, H. (2016). Leukocyte
opioid receptors mediate analgesia via Ca(2+)-regulated release of opioid peptides. Brain, Behavior & Immunity, 57, 227–242.
doi:10.1016/j.bbi.2016.04.018
Google Scholar WorldCat

Chan, H. C. S., McCarthy, D., Li, J., Palczewski, K., & Yuan, S. (2017). Designing safer analgesics via mu-opioid receptor pathways.
Trends in Pharmacological Sciences, 38(11), 1016–1037. doi:10.1016/j.tips.2017.08.004
Google Scholar WorldCat

Chidambaran, V., Sadhasivam, S., & Mahmoud, M. (2017). Codeine and opioid metabolism: Implications and alternatives for
pediatric pain management. Current Opinion in Anesthesiology, 30(3), 349–356. doi:10.1097/ACO.0000000000000455
Google Scholar WorldCat

Chou, R., Gordon, D. B., de Leon-Casasola, O. A., Rosenberg, J. M., Bickler, S., Brennan, T., … Wu, C. L. (2016). Management of
postoperative pain: A clinical practice guideline from the American Pain Society, the American Society of Regional Anesthesia
and Pain Medicine, and the American Society of Anesthesiologistsʼ Committee on Regional Anesthesia, Executive Committee,
and Administrative Council. Journal of Pain, 17(2), 131–157. doi:10.1016/j.jpain.2015.12.008
Google Scholar WorldCat

Christoph, A., Eerdekens, M. H., Kok, M., Volkers, G., & Freynhagen, R. (2017). Cebranopadol, a novel first-in-class analgesic drug
candidate: First experience in patients with chronic low back pain in a randomized clinical trial. Pain, 158(9), 1813–1824.
doi:10.1097/j.pain.0000000000000986
Google Scholar WorldCat

Codd, E. E., Carson, J. R., Colburn, R. W., Stone, D. J., Van Besien, C. R., Zhang, S. P., … Flores, C. M. (2009). JNJ-20788560 [9-(8-
p. 753 azabicyclo[3.2.1]oct-3-ylidene)-9H-xanthene-3-carboxylic acid diethylamide], a selective delta opioid receptor agonist, is a
potent and e icacious antihyperalgesic agent that does not produce respiratory depression, pharmacologic tolerance, or
physical dependence. Journal of Pharmacology & Experimental Therapeutics, 329(1), 241–251. doi:10.1124/jpet.108.146969

Collett, B. J. (1998). Opioid tolerance: The clinical perspective. British Journal of Anaesthesiology, 81(1), 58–68. Retrieved from
https://www.ncbi.nlm.nih.gov/pubmed/9771273
Google Scholar WorldCat

Collin, E., Poulain, P., Gauvain-Piquard, A., Petit, G., & Pichard-Leandri, E. (1993). Is disease progression the major factor in
morphine “tolerance” in cancer pain treatment? Pain, 55(3), 319–326. Retrieved from

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


http://www.ncbi.nlm.nih.gov/pubmed/8121693
Google Scholar WorldCat

Comelon, M., Raeder, J., Stubhaug, A., Nielsen, C. S., Draegni, T., & Lenz, H. (2016). Gradual withdrawal of remifentanil infusion
may prevent opioid-induced hyperalgesia. British Journal of Anaesthesiology, 116(4), 524–530. doi:10.1093/bja/aev547
Google Scholar WorldCat

Compton, P., Charuvastra, V. C., & Ling, W. (2001). Pain intolerance in opioid-maintained former opiate addicts: E ect of long-
acting maintenance agent. Drug & Alcohol Dependence, 63(2), 139–146. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/11376918
Google Scholar WorldCat

Compton, P., Miotto, K., & Elasho , D. (2004). Precipitated opioid withdrawal across acute physical dependence induction
methods. Pharmacology, Biochemistry, & Behavior, 77(2), 263–268. Retrieved from
http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=14751453
Google Scholar WorldCat

Corder, G., Tawfik, V. L., Wang, D., Sypek, E. I., Low, S. A., Dickinson, J. R., … Scherrer, G. (2017). Loss of mu opioid receptor
signaling in nociceptors, but not microglia, abrogates morphine tolerance without disrupting analgesia. Nature Medicine, 23(2),
164–173. doi:10.1038/nm.4262
Google Scholar WorldCat

Cummings, N. A., & Nordby, G. L. (1966). Measurement of synovial fluid pH in normal and arthritic knees. Arthritis & Rheumatism,
9(1), 47–56. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/4952418
Google Scholar WorldCat

Davis, K. D., Flor, H., Greely, H. T., Iannetti, G. D., Mackey, S., Ploner, M., … Wager, T. D. (2017). Brain imaging tests for chronic pain:
Medical, legal and ethical issues and recommendations. Nature Reviews Neurology, 13(10), 624–638.
doi:10.1038/nrneurol.2017.122
Google Scholar WorldCat

De Marco, R., Bedini, A., Spampinato, S., Cavina, L., Pirazzoli, E., & Gentilucci, L. (2016). Versatile picklocks to access all opioid
receptors: Tuning the selectivity and functional profile of the cyclotetrapeptide c[Phe-d-Pro-Phe-Trp] (CJ-15,208). Journal of
Medicinal Chemistry, 59(19), 9255–9261. doi:10.1021/acs.jmedchem.6b00420
Google Scholar WorldCat

Del Vecchio, G., Spahn, V., & Stein, C. (2017). Novel opioid analgesics and side e ects. ACS Chemical Neuroscience, 8(8), 1638–
1640. doi:10.1021/acschemneuro.7b00195
Google Scholar WorldCat

Dembla, S., Behrendt, M., Mohr, F., Goecke, C., Sondermann, J., Schneider, F. M., … Oberwinkler, J. (2017). Anti-nociceptive
action of peripheral mu-opioid receptors by G-beta-gamma protein-mediated inhibition of TRPM3 channels. Elife, 6.
doi:10.7554/eLife.26280
Google Scholar WorldCat
DeWire, S. M., Yamashita, D. S., Rominger, D. H., Liu, G., Cowan, C. L., Graczyk, T. M., … Violin, J. D. (2013). A G protein-biased
ligand at the mu-opioid receptor is potently analgesic with reduced gastrointestinal and respiratory dysfunction compared with
morphine. Journal of Pharmacology & Experimental Therapeutics, 344(3), 708–717. doi:10.1124/jpet.112.201616
Google Scholar WorldCat

Diener, H. C., Holle, D., Solbach, K., & Gaul, C. (2016). Medication-overuse headache: Risk factors, pathophysiology and
management. Nature Reviews Neurology, 12(10), 575–583. doi:10.1038/nrneurol.2016.124
Google Scholar WorldCat

Ding, H., Czoty, P. W., Kiguchi, N., Cami-Kobeci, G., Sukhtankar, D. D., Nader, M. A., … Ko, M. C. (2016). A novel orvinol analog,
p. 754 BU08028, as a safe opioid analgesic without abuse liability in primates. Proceedings of the National Academy of Sciences USA,

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


113(37), E5511–5518. doi:10.1073/pnas.1605295113

Drewes, A. M., Jensen, R. D., Nielsen, L. M., Droney, J., Christrup, L. L., Arendt-Nielsen, L., … Dahan, A. (2013). Di erences
between opioids: Pharmacological, experimental, clinical and economical perspectives. British Journal of Clinical
Pharmacology, 75(1), 60–78. doi:10.1111/j.1365-2125.2012.04317.x
Google Scholar WorldCat

Dzau, V. J., & Pizzo, P. A. (2014). Relieving pain in America: Insights from an Institute of Medicine committee. Journal of the
American Medical Association, 312(15), 1507–1508. doi:10.1001/jama.2014.12986
Google Scholar WorldCat

Elhabazi, K., Humbert, J. P., Bertin, I., Quillet, R., Utard, V., Schneider, S., … Simonin, F. (2017). RF313, an orally bioavailable
neuropeptide FF receptor antagonist, opposes e ects of RF-amide-related peptide-3 and opioid-induced hyperalgesia in
rodents. Neuropharmacology, 118, 188–198. doi:10.1016/j.neuropharm.2017.03.012
Google Scholar WorldCat

Endres-Becker, J., Heppenstall, P. A., Mousa, S. A., Labuz, D., Oksche, A., Schäfer, M., … Zöllner, C. (2007). Mu-opioid receptor
activation modulates transient receptor potential vanilloid 1 (TRPV1) currents in sensory neurons in a model of inflammatory
pain. Molecular Pharmacology, 71(1), 12–18. Retrieved from http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?
cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=17005903
Google Scholar WorldCat

Engel, C. C., von Kor , M., & Katon, W. J. (1996). Back pain in primary care: Predictors of high health-care costs. Pain, 65(2–3),
197–204. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/8826507
Google Scholar WorldCat

Eriksen, J., Sjogren, P., Bruera, E., Ekholm, O., & Rasmussen, N. K. (2006). Critical issues on opioids in chronic non-cancer pain: An
epidemiological study. Pain, 125(1–2), 172–179. Retrieved from http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?
cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=16842922
Google Scholar WorldCat

Falchuk, K. H., Goetzl, E. J., & Kulka, J. P. (1970). Respiratory gases of synovial fluids. An approach to synovial tissue circulatory-
metabolic imbalance in rheumatoid arthritis. American Journal of Medicine, 49(2), 223–231. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/5452943
Google Scholar WorldCat

Farr, M., Garvey, K., Bold, A. M., Kendall, M. J., & Bacon, P. A. (1985). Significance of the hydrogen ion concentration in synovial
fluid in rheumatoid arthritis. Clinical & Experimental Rheumatology, 3(2), 99–104. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/4017318
Google Scholar WorldCat

Ferrini, F., Trang, T., Mattioli, T. A., La ray, S., DelʼGuidice, T., Lorenzo, L. E., … De Koninck, Y. (2013). Morphine hyperalgesia gated
through microglia-mediated disruption of neuronal Cl(-) homeostasis. Nature Neuroscience, 16(2), 183–192. doi:10.1038/nn.3295
Google Scholar WorldCat

Fidecka, S., Malec, D., & Langwinski, R. (1978). Central action of narcotic analgesics. II. Locomotor activity and narcotic
analgesics. Polish Journal of Pharmacology & Pharmacy, 30(1), 5–16. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/25426
Google Scholar WorldCat

Fishbain, D. A., Cole, B., Lewis, J. E., Gao, J., & Rosomo , R. S. (2009). Do opioids induce hyperalgesia in humans? An evidence-
based structured review. Pain Medicine, 10(5), 829–839. doi:10.1111/j.1526-4637.2009.00653.x
Google Scholar WorldCat

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Fletcher, D., & Martinez, V. (2014). Opioid-induced hyperalgesia in patients a er surgery: A systematic review and a meta-
analysis. British Journal of Anaesthesiology, 112(6), 991–1004. doi:10.1093/bja/aeu137
Google Scholar WorldCat

Flor, H., & Diers, M. (2007). Limitations of pharmacotherapy: Behavioral approaches to chronic pain. Handbook of Experimental
Pharmacology, (177), 415–427. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/17087132
Google Scholar WorldCat

p. 755 Fordyce, W. E. (1991). Opioids and treatment targets. American Pain Society Bulletin, 1(4), 1–4.
Google Scholar WorldCat

Fordyce, W. E. (1992). Opioids, pain and behavioral outcomes. American Pain Society Journal, 1(4), 282–284.
Google Scholar WorldCat

Fordyce, W. E., Fowler, R. S., & DeLateur, B. (1968). An application of behavior modification technique to a problem of chronic
pain. Behaviour Research and Therapy, 6(1), 105–107. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/5644753
Google Scholar WorldCat

Fujita, W., Gomes, I., & Devi, L. A. (2015). Heteromers of mu-delta opioid receptors: new pharmacology and novel therapeutic
possibilities. British Journal of Pharmacology, 172(2), 375–387. doi:10.1111/bph.12663
Google Scholar WorldCat

Galer, B. S., Lee, D., Ma, T., Nagle, B., & Schlagheck, T. G. (2005). MorphiDex (morphine sulfate/dextromethorphan hydrobromide
combination) in the treatment of chronic pain: Three multicenter, randomized, double-blind, controlled clinical trials fail to
demonstrate enhanced opioid analgesia or reduction in tolerance. Pain, 115(3), 284–295. doi:10.1016/j.pain.2005.03.004
Google Scholar WorldCat

Gallagher, F. A., Kettunen, M. I., Day, S. E., Hu, D. E., Ardenkjaer-Larsen, J. H., Zandt, R., … Brindle, K. M. (2008). Magnetic
resonance imaging of pH in vivo using hyperpolarized 13C-labelled bicarbonate. Nature, 453(7197), 940–943.
doi:10.1038/nature07017
Google Scholar WorldCat

Gatchel, R. J., & Okifuji, A. (2006). Evidence-based scientific data documenting the treatment and cost-e ectiveness of
comprehensive pain programs for chronic nonmalignant pain. Journal of Pain, 7(11), 779–793. doi:10.1016/j.jpain.2006.08.005
Google Scholar WorldCat

Gaveriaux-Ru , C. (2013). Opiate-induced analgesia: Contributions from mu, delta and kappa opioid receptors mouse mutants.
Current Pharmaceutical Design, 19(42), 7373–7381. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/23448470
Google Scholar WorldCat

Gaveriaux-Ru , C., Karchewski, L. A., Hever, X., Matifas, A., & Kie er, B. L. (2008). Inflammatory pain is enhanced in delta opioid
receptor-knockout mice. European Journal of Neuroscience, 27(10), 2558–2567. doi:10.1111/j.1460-9568.2008.06223.x
Google Scholar WorldCat
Gaveriaux-Ru , C., & Kie er, B. L. (2011). Delta opioid receptor analgesia: Recent contributions from pharmacology and
molecular approaches. Behavioral Pharmacology, 22(5–6), 405–414. doi:10.1097/FBP.0b013e32834a1f2c
Google Scholar WorldCat

Geborek, P., Saxne, T., Pettersson, H., & Wollheim, F. A. (1989). Synovial fluid acidosis correlates with radiological joint
destruction in rheumatoid arthritis knee joints. Journal of Rheumatology, 16(4), 468–472. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/2746586
Google Scholar WorldCat

Gendron, L., Cahill, C. M., von Zastrow, M., Schiller, P. W., & Pineyro, G. (2016). Molecular pharmacology of delta-opioid receptors.
Pharmacological Reviews, 68(3), 631–700. doi:10.1124/pr.114.008979

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Google Scholar WorldCat

Gillies, R. J., Liu, Z., & Bhujwalla, Z. (1994). 31P-MRS measurements of extracellular pH of tumors using 3-
aminopropylphosphonate. The American Journal of Physiology, 267(1 Pt 1), C195–C203. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/8048479
Google Scholar WorldCat

Goins, W. F., Cohen, J. B., & Glorioso, J. C. (2012). Gene therapy for the treatment of chronic peripheral nervous system pain.
Neurobiology of Disease, 48(2), 255–270. doi:10.1016/j.nbd.2012.05.005
Google Scholar WorldCat

Goldie, I., & Nachemson, A. (1969). Synovial pH in rheumatoid knee-joints. I. The e ect of synovectomy. Acta Orthopaedica
Scandinavica, 40(5), 634–641. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/5378127
Google Scholar WorldCat

Goldie, I., & Nachemson, A. (1970). Synovial pH in rheumatoid knee joints. II. The e ect of local corticosteroid treatment. Acta
Orthopaedica Scandinavica, 41(3), 354–362. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/5486188
Google Scholar WorldCat

p. 756 Gomes, I., Ayoub, M. A., Fujita, W., Jaeger, W. C., Pfleger, K. D., & Devi, L. A. (2016). G protein-coupled receptor heteromers. Annual
Review of Pharmacology & Toxicology, 56, 403–425. doi:10.1146/annurev-pharmtox-011613-135952
Google Scholar WorldCat

Gonzalez-Rodriguez, S., Quadir, M. A., Gupta, S., Walker, K. A., Zhang, X., Spahn, V., … Stein, C. (2017). Polyglycerol-opioid
conjugate produces analgesia devoid of side e ects. Elife, 6, e27081. doi:10.7554/eLife.27081
Google Scholar WorldCat

Granier, S., Manglik, A., Kruse, A. C., Kobilka, T. S., Thian, F. S., Weis, W. I., & Kobilka, B. K. (2012). Structure of the delta-opioid
receptor bound to naltrindole. Nature, 485(7398), 400–404. doi:10.1038/nature11111
Google Scholar WorldCat

Grenald, S. A., Young, M. A., Wang, Y., Ossipov, M. H., Ibrahim, M. M., Largent-Milnes, T. M., & Vanderah, T. W. (2017). Synergistic
attenuation of chronic pain using mu opioid and cannabinoid receptor 2 agonists. Neuropharmacology, 116, 59–70.
doi:10.1016/j.neuropharm.2016.12.008
Google Scholar WorldCat

Gri in, M. L., McDermott, K. A., McHugh, R. K., Fitzmaurice, G. M., Jamison, R. N., & Weiss, R. D. (2016). Longitudinal association
between pain severity and subsequent opioid use in prescription opioid dependent patients with chronic pain. Drug & Alcohol
Dependence, 163, 216–221. doi:10.1016/j.drugalcdep.2016.04.023
Google Scholar WorldCat

Guedon, J. M., Wu, S., Zheng, X., Churchill, C. C., Glorioso, J. C., Liu, C. H., … Hao, S. (2015). Current gene therapy using viral
vectors for chronic pain. Molecular Pain, 11, 27. doi:10.1186/s12990-015-0018-1
Google Scholar WorldCat
Gunther, T., Dasgupta, P., Mann, A., Miess, E., Kliewer, A., Fritzwanker, S., … Schulz, S. (2017). Targeting multiple opioid receptors
—improved analgesics with reduced side e ects? British Journal of Pharmacology. doi:10.1111/bph.13809
Google Scholar WorldCat

Gustavsson, A., Bjorkman, J., Ljungcrantz, C., Rhodin, A., Rivano-Fischer, M., Sjolund, K. F., & Mannheimer, C. (2012).
Pharmaceutical treatment patterns for patients with a diagnosis related to chronic pain initiating a slow-release strong opioid
treatment in Sweden. Pain, 153(12), 2325–2331. doi:10.1016/j.pain.2012.07.011
Google Scholar WorldCat

Häbler, C. (1929). Über den K- und Ca-Gehalt von Eiter und Exsudaten und seine Beziehungen zum Entzündungsschmerz.
Klinische Wochenschri , 34, 1569–1572.

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Google Scholar WorldCat

Hassan, A. H. S., Ableitner, A., Stein, C., & Herz, A. (1993). Inflammation of the rat paw enhances axonal transport of opioid
receptors in the sciatic nerve and increases their density in the inflamed tissue. Neuroscience, 55, 185–195.
Google Scholar WorldCat

Hauser, A. S., Chavali, S., Masuho, I., Jahn, L. J., Martemyanov, K. A., Gloriam, D. E., & Babu, M. M. (2018). Pharmacogenomics of
GPCR drug targets. Cell, 172(1–2), 41–54, e19. doi:10.1016/j.cell.2017.11.033
Google Scholar WorldCat

Healy, J. R., Bezawada, P., Griggs, N. W., Devereaux, A. L., Matsumoto, R. R., Traynor, J. R., … Cunningham, C. W. (2017).
Benzylideneoxymorphone: A new lead for development of bifunctional mu/delta opioid receptor ligands. Bioorganic & Medicinal
Chemistry Letters, 27(3), 666–669. doi:10.1016/j.bmcl.2016.11.057
Google Scholar WorldCat

Herz, A., Millan, M. J., & Stein, C. (1989). Arthritic inflammation in rats as a model of chronic pain: Role of opioid systems. NIDA
Research Monograph, 95, 110–115. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/2561818
Google Scholar WorldCat

Holmes, R. D., Tiwari, A. K., & Kennedy, J. L. (2016). Mechanisms of the placebo e ect in pain and psychiatric disorders.
Pharmacogenomics Journal, 16(6), 491–500. doi:10.1038/tpj.2016.15
Google Scholar WorldCat

Hood, V. L., Schubert, C., Keller, U., & Muller, S. (1988). E ect of systemic pH on pHi and lactic acid generation in exhaustive
forearm exercise. American Journal of Physiology, 255(3 Pt 2), F479–F485. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/3414804
Google Scholar WorldCat

p. 757 Hua, Z., Liu, L., Shen, J., Cheng, K., Liu, A., Yang, J., … Cheng, J. (2016). Mesenchymal stem cells reversed morphine tolerance and
opioid-induced hyperalgesia. Scientific Reports, 6, 32096. doi:10.1038/srep32096
Google Scholar WorldCat

Huang, P., Chen, C., & Liu-Chen, L. Y. (2015). Detection of mu opioid receptor (MOPR) and its glycosylation in rat and mouse brains
by western blot with anti-muC, an a inity-purified polyclonal anti-MOPR antibody. Methods in Molecular Biology, 1230, 141–154.
doi:10.1007/978-1-4939-1708-2_11
Google Scholar WorldCat

Hubner, H., Schellhorn, T., Gienger, M., Schaab, C., Kaindl, J., Leeb, L., … Gmeiner, P. (2016). Structure-guided development of
heterodimer-selective GPCR ligands. Nature Communications, 7, 12298. doi:10.1038/ncomms12298
Google Scholar WorldCat

Hutchins, G. M., & Sheldon, W. H. (1972). The pH of inflammatory exudates in acidotic diabetic rabbits. Proceedings of the Society
for Experimental Biology & Medicine, 140(2), 623–627. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/5037600
Google Scholar WorldCat
Hutchinson, M. R., Shavit, Y., Grace, P. M., Rice, K. C., Maier, S. F., & Watkins, L. R. (2011). Exploring the
neuroimmunopharmacology of opioids: An integrative review of mechanisms of central immune signaling and their implications
for opioid analgesia. Pharmacological Reviews, 63(3), 772–810. doi:10.1124/pr.110.004135
Google Scholar WorldCat

Imam, M. Z., Kuo, A., Ghassabian, S., & Smith, M. T. (2017). Progress in understanding mechanisms of opioid-induced
gastrointestinal adverse e ects and respiratory depression. Neuropharmacology, 131, 238–255.
doi:10.1016/j.neuropharm.2017.12.032
Google Scholar WorldCat

Issberner, U., Reeh, P. W., & Steen, K. H. (1996). Pain due to tissue acidosis: A mechanism for inflammatory and ischemic myalgia?

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Neuroscience Letters, 208(3), 191–194. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/8733302
Google Scholar WorldCat

Jacobus, W. E., Taylor, G. J. t., Hollis, D. P., & Nunnally, R. L. (1977). Phosphorus nuclear magnetic resonance of perfused working
rat hearts. Nature, 265(5596), 756–758. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/16217
Google Scholar WorldCat

Jamshidi, R. J., Jacobs, B. A., Sullivan, L. C., Chavera, T. A., Saylor, R. M., Prisinzano, T. E., … Berg, K. A. (2015). Functional
selectivity of kappa opioid receptor agonists in peripheral sensory neurons. Journal of Pharmacology & Experimental
Therapeutics, 355(2), 174–182. doi:10.1124/jpet.115.225896
Google Scholar WorldCat

Javelot, H., Messaoudi, M., Garnier, S., & Rougeot, C. (2010). Human opiorphin is a naturally occurring antidepressant acting
selectively on enkephalin-dependent delta-opioid pathways. Journal of Physiology & Pharmacology, 61(3), 355–362. Retrieved
from https://www.ncbi.nlm.nih.gov/pubmed/20610867
Google Scholar WorldCat

Jebens, E. H., & Monk-Jones, M. E. (1959). On the viscosity and pH of synovial fluid and the pH of blood. Journal of Bone & Joint
Surgery (British), 41-B(2), 388–400. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/13641329
Google Scholar WorldCat

Kager, I., Mousa, S. A., Sieper, J., Stein, C., Pipam, W., & Likar, R. (2011). Blockade of intra-articular adrenergic receptors increases
analgesic demands for pain relief a er knee surgery. Rheumatology International, 31(10), 1299–1306. doi:10.1007/s00296-010-
1489-z
Google Scholar WorldCat

Kie er, B. L. (2016). Drug discovery: Designing the ideal opioid. Nature, 537(7619), 170–171. doi:10.1038/nature19424
Google Scholar WorldCat

Kie er, B. L., & Gaveriaux-Ru , C. (2002). Exploring the opioid system by gene knockout. Progress in Neurobiology, 66(5), 285–
306. Retrieved from https://www.ncbi.nlm.nih.gov/pubmed/12015197
Google Scholar WorldCat

Klinck, M. P., Mogil, J. S., Moreau, M., Lascelles, B. D. X., Flecknell, P. A., Poitte, T., & Troncy, E. (2017). Translational pain
assessment: Could natural animal models be the missing link? Pain, 158(9), 1633–1646. doi:10.1097/j.pain.0000000000000978
Google Scholar WorldCat

p. 758 Koblish, M., Carr, R., 3rd, Siuda, E. R., Rominger, D. H., Gowen-MacDonald, W., Cowan, C. L., … Lark, M. W. (2017). TRV0109101, a G
protein-biased agonist of the micro-opioid receptor, does not promote opioid-induced mechanical allodynia following chronic
administration. Journal of Pharmacology & Experimental Therapeutics, 362(2), 254–262. doi:10.1124/jpet.117.241117
Google Scholar WorldCat

Koldajew, B., & Altschuler, M. (1930). Zur Frage über die aktive Reaktion der Exsudate bei chronischen und akuten Entzündungen.
Zeitschri für Immunitätsforschung, 69, 18–24.
Google Scholar WorldCat

Kringel, D., Ultsch, A., Zimmermann, M., Jansen, J. P., Ilias, W., Freynhagen, R., … Lötsch, J. (2017). Emergent biomarker derived
from next-generation sequencing to identify pain patients requiring uncommonly high opioid doses. Pharmacogenomics
Journal, 17, 419–426. doi:10.1038/tpj.2016.28
Google Scholar WorldCat

Kruegel, A. C., Gassaway, M. M., Kapoor, A., Varadi, A., Majumdar, S., Filizola, M., … Sames, D. (2016). Synthetic and receptor
signaling explorations of the mitragyna alkaloids: Mitragynine as an atypical molecular framework for opioid receptor
modulators. Journal of the American Chemical Society, 138(21), 6754–6764. doi:10.1021/jacs.6b00360
Google Scholar WorldCat

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Labuz, D., Celik, M. O., Zimmer, A., & Machelska, H. (2016). Distinct roles of exogenous opioid agonists and endogenous opioid
peptides in the peripheral control of neuropathy-triggered heat pain. Scientific Reports, 6, 32799. doi:10.1038/srep32799
Google Scholar WorldCat

Labuz, D., Mousa, S. A., Schäfer, M., Stein, C., & Machelska, H. (2007). Relative contribution of peripheral versus central opioid
receptors to anti-nociception. Brain Research, 1160, 30–38. Retrieved from http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?
cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=17599812
Google Scholar WorldCat

Labuz, D., Schmidt, Y., Schreiter, A., Rittner, H. L., Mousa, S. A., & Machelska, H. (2009). Immune cell-derived opioids protect
against neuropathic pain in mice. Journal of Clinical Investigation, 119(2), 278–286. doi:36246 [pii] 10.1172/JCI36246
Google Scholar WorldCat

Lagard, C., Chevillard, L., Guillemyn, K., Risede, P., Laplanche, J. L., Spetea, M., … Megarbane, B. (2017). Bifunctional peptide-
based opioid agonist/nociceptin antagonist ligand for dual treatment of nociceptive and neuropathic pain. Pain, 158(3), 505–
515. doi:10.1097/j.pain.0000000000000790
Google Scholar WorldCat

Lamusuo, S., Hirvonen, J., Lindholm, P., Martikainen, I. K., Hagelberg, N., Parkkola, R., … Jaaskelainen, S. K. (2017).
Neurotransmitters behind pain relief with transcranial magnetic stimulation—positron emission tomography evidence for
release of endogenous opioids. European Journal of Pain, 21(9), 1505–1515. doi:10.1002/ejp.1052
Google Scholar WorldCat

Langford, R. M., Knaggs, R., Farquhar-Smith, P., & Dickenson, A. H. (2016). Is tapentadol di erent from classical opioids? A review
of the evidence. British Journal of Pain, 10(4), 217–221. doi:10.1177/2049463716657363
Google Scholar WorldCat

Larsson, M. H., Bayati, A., Lindstrom, E., & Larsson, H. (2008). Involvement of kappa-opioid receptors in visceral nociception in
mice. Neurogastroenterology & Motility, 20(10), 1157–1164. doi:10.1111/j.1365-2982.2008.01161.x
Google Scholar WorldCat

Law, P. Y., Reggio, P. H., & Loh, H. H. (2013). Opioid receptors: Toward separation of analgesic from undesirable e ects. Trends in
Biochemical Sciences, 38(6), 275–282. doi:10.1016/j.tibs.2013.03.003
Google Scholar WorldCat

Le Naour, M., Lunzer, M. M., Powers, M. D., Kalyuzhny, A. E., Benneyworth, M. A., Thomas, M. J., & Portoghese, P. S. (2014).
Putative kappa opioid heteromers as targets for developing analgesics free of adverse e ects. Journal of Medicinal Chemistry,
57(15), 6383–6392. doi:10.1021/jm500159d
Google Scholar WorldCat

p. 759 Leadley, R. M., Armstrong, N., Lee, Y. C., Allen, A., & Kleijnen, J. (2012). Chronic diseases in the European Union: The prevalence
and health cost implications of chronic pain. Journal of Pain & Palliative Care Pharmacotherapy, 26(4), 310–325.
doi:10.3109/15360288.2012.736933
Google Scholar WorldCat

Lee, M. C., Wanigasekera, V., & Tracey, I. (2012). Imaging opioid analgesia in the human brain. Trends in Anaesthesiology & Critical
Care, 2, 244–248.
Google Scholar WorldCat

Legrain, V., Iannetti, G. D., Plaghki, L., & Mouraux, A. (2011). The pain matrix reloaded: A salience detection system for the body.
Progress in Neurobiology, 93(1), 111–124. doi:10.1016/j.pneurobio.2010.10.005
Google Scholar WorldCat

Levine, N., & Kelly, H. (1978). Measurement of pH in the rat epididymis in vivo. Journal of Reproductive Fertility, 52(2), 333–335.

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/24738
Google Scholar WorldCat

Likar, R., Mousa, S. A., Steinkellner, H., Koppert, W., Philippitsch, G., Stein, C., & Schäfer, M. (2007). Involvement of intraarticular
corticotropin-releasing hormone in postoperative pain modulation. Clinical Journal of Pain, 23(2), 136–142. Retrieved from
http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=17237662
Google Scholar WorldCat

Livingston, K. E., & Traynor, J. R. (2017). Allostery at opioid receptors: Modulation with small molecule ligands. British Journal of
Pharmacology. doi:10.1111/bph.13823
Google Scholar WorldCat

Loeser, J. D., & Treede, R. D. (2008). The Kyoto protocol of IASP basic pain terminology. Pain, 137(3), 473–477. Retrieved from
http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=18583048
Google Scholar WorldCat

Machelska, H. (2011). Dual peripheral actions of immune cells in neuropathic pain. Archivum Immunologiae et Therapiae
Experimentalis, 59(1), 11–24. doi:10.1007/s00005-010-0106-x
Google Scholar WorldCat

Madariaga-Mazon, A., Marmolejo-Valencia, A. F., Li, Y., Toll, L., Houghten, R. A., & Martinez-Mayorga, K. (2017). Mu-opioid receptor
biased ligands: A safer and painless discovery of analgesics? Drug Discovery Today, 22(11), 1719–1729.
doi:10.1016/j.drudis.2017.07.002
Google Scholar WorldCat

Madasu, M. K., Roche, M., & Finn, D. P. (2015). Supraspinal transient receptor potential subfamily V member 1 (TRPV1) in pain and
psychiatric disorders. Modern Trends in Pharmacopsychiatry, 30, 80–93. doi:10.1159/000435934
Google Scholar WorldCat

Maddila, S. C., Busch-Dienstfertig, M., & Stein, C. (2017). B lymphocytes express POMC mRNA, processing enzymes and beta-
endorphin in painful inflammation. Journal of Neuroimmune Pharmacology, 12(1), 180–186. doi:10.1007/s11481-016-9715-4
Google Scholar WorldCat

Maillet, E. L., Milon, N., Heghinian, M. D., Fishback, J., Schurer, S. C., Garamszegi, N., & Mash, D. C. (2015). Noribogaine is a G-
protein biased kappa-opioid receptor agonist. Neuropharmacology, 99, 675–688. doi:10.1016/j.neuropharm.2015.08.032
Google Scholar WorldCat

Manglik, A., Kruse, A. C., Kobilka, T. S., Thian, F. S., Mathiesen, J. M., Sunahara, R. K., … Granier, S. (2012). Crystal structure of the
mu-opioid receptor bound to a morphinan antagonist. Nature, 485(7398), 321–326.
doi:http://www.nature.com/nature/journal/v485/n7398/abs/nature10954.html#supplementary-information
Google Scholar WorldCat

Manglik, A., Lin, H., Aryal, D. K., McCorvy, J. D., Dengler, D., Corder, G., … Shoichet, B. K. (2016). Structure-based discovery of
opioid analgesics with reduced side e ects. Nature, 537(7619), 185–190. doi:10.1038/nature19112
Google Scholar WorldCat

Mansikka, H., Zhao, C., Sheth, R. N., Sora, I., Uhl, G., & Raja, S. N. (2004). Nerve injury induces a tonic bilateral mu-opioid receptor-
mediated inhibitory e ect on mechanical allodynia in mice. Anesthesiology, 100(4), 912–921. Retrieved from
https://www.ncbi.nlm.nih.gov/pubmed/15087627
Google Scholar WorldCat

p. 760 Mansikka, H., Zhou, L., Donovan, D. M., Pertovaara, A., & Raja, S. N. (2002). The role of mu-opioid receptors in inflammatory
hyperalgesia and alpha 2-adrenoceptor-mediated antihyperalgesia. Neuroscience, 113(2), 339–349. Retrieved from
https://www.ncbi.nlm.nih.gov/pubmed/12127091
Google Scholar WorldCat

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Martins, D., Tavares, I., & Morgado, C. (2014). “Hotheaded”: The role of TRPV1 in brain functions. Neuropharmacology, 85, 151–
157. doi:10.1016/j.neuropharm.2014.05.034
Google Scholar WorldCat

Massotte, D. (2015). In vivo opioid receptor heteromerization: Where do we stand? British Journal of Pharmacology, 172(2), 420–
434. doi:10.1111/bph.12702
Google Scholar WorldCat

Matic, M., de Wildt, S. N., Tibboel, D., & van Schaik, R. H. N. (2017). Analgesia and opioids: A pharmacogenetics shortlist for
implementation in clinical practice. Clinical Chemistry, 63(7), 1204–1213. doi:10.1373/clinchem.2016.264986
Google Scholar WorldCat

Mauermann, E., Filitz, J., Dolder, P., Rentsch, K. M., Bandschapp, O., & Ruppen, W. (2016). Does fentanyl lead to opioid-induced
hyperalgesia in healthy volunteers? A double-blind, randomized, crossover trial. Anesthesiology, 124(2), 453–463.
doi:10.1097/ALN.0000000000000976
Google Scholar WorldCat

McNicol, E. (2008). Opioid side e ects and their treatment in patients with chronic cancer and noncancer pain. Journal of Pain &
Palliative Care Pharmacotherapy, 22(4), 270–281. doi:10.1080/15360280802537225
Google Scholar WorldCat

Mechling, A. E., Arefin, T., Lee, H. L., Bienert, T., Reisert, M., Ben Hamida, S., … Harsan, L. A. (2016). Deletion of the mu opioid
receptor gene in mice reshapes the reward-aversion connectome. Proceedings of the National Academy of Sciences USA, 113(41),
11603–11608. doi:10.1073/pnas.1601640113
Google Scholar WorldCat

Melik Parsadaniantz, S., Rivat, C., Rostene, W., & Reaux-Le Goazigo, A. (2015). Opioid and chemokine receptor crosstalk: A
promising target for pain therapy? Nature Reviews Neuroscience, 16(2), 69–78. doi:10.1038/nrn3858
Google Scholar WorldCat

Menkin, V. (1934). Studies on Inflammation: X. The cytological picture of an inflammatory exudate in relation to its hydrogen ion
concentration. American Journal of Pathology, 10(2), 193–210. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/19970136
Google Scholar WorldCat

Menkin, V. (1956). The role of hydrogen ion concentration and the cytology of an exudate. In C. C. Thomas (Ed.), Biochemical
Mechanisms in Inflammation (pp. 66–103). Springfield, IL: Charles C. Thomas Publisher Ltd.
Google Scholar Google Preview WorldCat COPAC

Menkin, V., & Warner, C. R. (1937). Studies on inflammation: XIII. Carbohydrate metabolism, local acidosis, and the cytological
picture in inflammation. American Journal of Pathology, 13(1), 25–44 21. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/19970311
Google Scholar WorldCat
Mennini, N., Mura, P., Nativi, C., Richichi, B., Di Cesare Mannelli, L., & Ghelardini, C. (2015). Injectable liposomal formulations of
opiorphin as a new therapeutic strategy in pain management. Future Science OA, 1(3), FSO2. doi:10.4155/fso.14.3
Google Scholar WorldCat

Meyer, K. A., Kammerling, E. M., et al. (1948). pH studies of malignant tissues in human beings. Cancer Research, 8(11), 513–518.
Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/18114935
Google Scholar WorldCat

Minett, M. S., Pereira, V., Sikandar, S., Matsuyama, A., Lolignier, S., Kanellopoulos, A. H., … Wood, J. N. (2015). Endogenous
opioids contribute to insensitivity to pain in humans and mice lacking sodium channel Nav1.7. Nature Communications, 6, 8967.
doi:10.1038/ncomms9967

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Google Scholar WorldCat

Mogil, J. S., Davis, K. D., & Derbyshire, S. W. (2010). The necessity of animal models in pain research. Pain, 151(1), 12–17.
doi:10.1016/j.pain.2010.07.015
Google Scholar WorldCat

p. 761 Mollica, A., Carotenuto, A., Novellino, E., Limatola, A., Costante, R., Pinnen, F., … Hruby, V. J. (2014). Novel cyclic biphalin
analogue with improved anti-nociceptive properties. ACS Medicinal Chemistry Letters, 5(9), 1032–1036. doi:10.1021/ml500241n
Google Scholar WorldCat

Mousa, S. A., Cheppudira, B. P., Shaqura, M., Fischer, O., Hofmann, J., Hellweg, R., & Schafer, M. (2007). Nerve growth factor
governs the enhanced ability of opioids to suppress inflammatory pain. Brain, 130(Pt 2), 502–513. Retrieved from
http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=17142830
Google Scholar WorldCat

Mousa, S. A., Shakibaei, M., Sitte, N., Schäfer, M., & Stein, C. (2004). Subcellular pathways of beta-endorphin synthesis,
processing, and release from immunocytes in inflammatory pain. Endocrinology, 145(3), 1331–1341.
Google Scholar WorldCat

Mousa, S. A., Straub, R. H., Schäfer, M., & Stein, C. (2007). Beta-endorphin, met-enkephalin and corresponding opioid receptors
within synovium of patients with joint trauma, osteoarthritis and rheumatoid arthritis. Annals of the Rheumatic Diseases, 66(7),
871–879. Retrieved from http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?
cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=17324971
Google Scholar WorldCat

Mura, E., Govoni, S., Racchi, M., Carossa, V., Ranzani, G. N., Allegri, M., & van Schaik, R. H. (2013). Consequences of the 118A>G
polymorphism in the OPRM1 gene: Translation from bench to bedside? Journal of Pain Research, 6, 331–353.
doi:10.2147/JPR.S42040
Google Scholar WorldCat

Nadal, X., Banos, J. E., Kie er, B. L., & Maldonado, R. (2006). Neuropathic pain is enhanced in delta-opioid receptor knockout
mice. European Journal of Neuroscience, 23(3), 830–834. doi:10.1111/j.1460-9568.2006.04569.x
Google Scholar WorldCat

Newell, K., Franchi, A., Pouyssegur, J., & Tannock, I. (1993). Studies with glycolysis-deficient cells suggest that production of
lactic acid is not the only cause of tumor acidity. Proceedings of the National Academy of Sciences USA, 90(3), 1127–1131.
Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/8430084
Google Scholar WorldCat

Noble, F., & Cox, B. M. (1995). Di erential regulation of D1 dopamine receptor- and of A2a adenosine receptor-stimulated
adenylyl cyclase by mu-, delta 1-, and delta 2-opioid agonists in rat caudate putamen. Journal of Neurochemistry, 65(1), 125–133.
Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/7790855
Google Scholar WorldCat
Noble, F., & Cox, B. M. (1996). Di erences among mouse strains in the regulation by mu, delta 1 and delta 2 opioid receptors of
striatal adenylyl cyclases activated by dopamine D1 or adenosine A2a receptors. Brain Research, 716(1–2), 107–117.
doi:10.1016/0006-8993(96)00005-4
Google Scholar WorldCat

Noble, M., Treadwell, J. R., Tregear, S. J., Coates, V. H., Wi en, P. J., Akafomo, C., & Schoelles, K. M. (2010). Long-term opioid
management for chronic noncancer pain. Cochrane Database of Systematic Reviews (1), CD006605.
doi:10.1002/14651858.CD006605.pub2
Google Scholar WorldCat

Nockemann, D., Rouault, M., Labuz, D., Hublitz, P., McKnelly, K., Reis, F. C., … Heppenstall, P. A. (2013). The K channel GIRK2 is

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


both necessary and su icient for peripheral opioid-mediated analgesia. EMBO Molecular Medicine, 5(8), 1263–1277.
doi:10.1002/emmm.201201980
Google Scholar WorldCat

Nozaki, C., Le Bourdonnec, B., Reiss, D., Windh, R. T., Little, P. J., Dolle, R. E., … Gaveriaux-Ru , C. (2012). Delta-opioid
mechanisms for ADL5747 and ADL5859 e ects in mice: Analgesia, locomotion, and receptor internalization. Journal of
Pharmacology & Experimental Therapeutics, 342(3), 799–807. doi:10.1124/jpet.111.188987
Google Scholar WorldCat

Nozaki, C., Nagase, H., Nemoto, T., Matifas, A., Kie er, B. L., & Gaveriaux-Ru , C. (2014). In vivo properties of KNT-127, a novel
delta opioid receptor agonist: Receptor internalization, antihyperalgesia and antidepressant e ects in mice. British Journal of
Pharmacology, 171(23), 5376–5386. doi:10.1111/bph.12852
Google Scholar WorldCat

p. 762 Ojugo, A. S., McSheehy, P. M., McIntyre, D. J., McCoy, C., Stubbs, M., Leach, M. O., … Gri iths, J. R. (1999). Measurement of the
extracellular pH of solid tumours in mice by magnetic resonance spectroscopy: A comparison of exogenous (19)F and (31)P
probes. NMR in Biomedicine, 12(8), 495–504. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/10668042
Google Scholar WorldCat

Olson, K. M., Lei, W., Keresztes, A., LaVigne, J., & Streicher, J. M. (2017). Novel molecular strategies and targets for opioid drug
discovery for the treatment of chronic pain. Yale Journal of Biology & Medicine, 90(1), 97–110. Retrieved from
https://www.ncbi.nlm.nih.gov/pubmed/28356897
Google Scholar WorldCat

Pampus, F. (1963). [Hydrogen-ion concentration of brain tissue in space-occupying intracranial processes]. Acta Neurochirurgica
(Wien), 11, 305–318. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/14059253
Google Scholar WorldCat

Pan, J. W., Hamm, J. R., Rothman, D. L., & Shulman, R. G. (1988). Intracellular pH in human skeletal muscle by 1H NMR.
Proceedings of the National Academy of Sciences USA, 85(21), 7836–7839. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/3186694
Google Scholar WorldCat

Pannell, M., Labuz, D., Celik, M. O., Keye, J., Batra, A., Siegmund, B., & Machelska, H. (2016). Adoptive transfer of M2 macrophages
reduces neuropathic pain via opioid peptides. Journal of Neuroinflammation, 13(1), 262. doi:10.1186/s12974-016-0735-z
Google Scholar WorldCat

Pasternak, G. W., & Pan, Y. X. (2013). Mu opioids and their receptors: Evolution of a concept. Pharmacological Reviews, 65(4),
1257–1317. doi:10.1124/pr.112.007138
Google Scholar WorldCat

Peppin, J. F., & Ra a, R. B. (2015). Delta opioid agonists: A concise update on potential therapeutic applications. Journal of
Clinical Pharmacy & Therapeutics, 40(2), 155–166. doi:10.1111/jcpt.12244
Google Scholar WorldCat
Piomelli, D., & Sasso, O. (2014). Peripheral gating of pain signals by endogenous lipid mediators. Nature Neuroscience, 17(2),
164–174. doi:10.1038/nn.3612
Google Scholar WorldCat

Popik, P., Kamysz, E., Kreczko, J., & Wrobel, M. (2010). Human opiorphin: The lack of physiological dependence, tolerance to anti-
nociceptive e ects and abuse liability in laboratory mice. Behavioural Brain Research, 213(1), 88–93.
doi:10.1016/j.bbr.2010.04.045
Google Scholar WorldCat

Poras, H., Bonnard, E., Fournie-Zaluski, M. C., & Roques, B. P. (2015). Modulation of disulfide dual ENKephalinase inhibitors
(DENKIs) activity by a transient N-protection for pain alleviation by oral route. European Journal of Medicinal Chemistry, 102, 58–

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


67. doi:10.1016/j.ejmech.2015.07.027
Google Scholar WorldCat

Pradhan, A. A., Befort, K., Nozaki, C., Gaveriaux-Ru , C., & Kie er, B. L. (2011). The delta opioid receptor: An evolving target for the
treatment of brain disorders. Trends in Pharmacological Sciences, 32(10), 581–590. doi:10.1016/j.tips.2011.06.008
Google Scholar WorldCat

Pradhan, A. A., Perroy, J., Walwyn, W. M., Smith, M. L., Vicente-Sanchez, A., Segura, L., … Evans, C. J. (2016). Agonist-specific
recruitment of arrestin isoforms di erentially modify delta opioid receptor function. Journal of Neuroscience, 36(12), 3541–3551.
doi:10.1523/JNEUROSCI.4124-15.2016
Google Scholar WorldCat

Pradhan, A. A., Smith, M. L., Kie er, B. L., & Evans, C. J. (2012). Ligand-directed signalling within the opioid receptor family.
British Journal of Pharmacology, 167(5), 960–969. doi:10.1111/j.1476-5381.2012.02075.x
Google Scholar WorldCat

Prezzavento, O., Arena, E., Sanchez-Fernandez, C., Turnaturi, R., Parenti, C., Marrazzo, A., … Cobos, E. J. (2017). (+)-and (–)-
Phenazocine enantiomers: Evaluation of their dual opioid agonist/sigma1 antagonist properties and anti-nociceptive e ects.
European Journal of Medicinal Chemistry, 125, 603–610. doi:10.1016/j.ejmech.2016.09.077
Google Scholar WorldCat

Psaty, B. M., & Merrill, J. O. (2017). Addressing the opioid epidemic—Opportunities in the postmarketing setting. New England
Journal of Medicine, 376(16), 1502–1504. doi:10.1056/NEJMp1614972
Google Scholar WorldCat

p. 763 Punnia-Moorthy, A. (1987). Evaluation of pH changes in inflammation of the subcutaneous air pouch lining in the rat, induced by
carrageenan, dextran and Staphylococcus aureus. Journal of Oral Pathology, 16(1), 36–44. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/2435877

Quallo, T., Alkhatib, O., Gentry, C., Andersson, D. A., & Bevan, S. (2017). G protein betagamma subunits inhibit TRPM3 ion
channels in sensory neurons. Elife, 6. doi:10.7554/eLife.26138
Google Scholar WorldCat

Raehal, K. M., Schmid, C. L., Groer, C. E., & Bohn, L. M. (2011). Functional selectivity at the mu-opioid receptor: Implications for
understanding opioid analgesia and tolerance. Pharmacological Reviews, 63(4), 1001–1019. doi:10.1124/pr.111.004598
Google Scholar WorldCat

Ranjan, R., Pandey, S., & Shukla, A. K. (2017). Biased opioid receptor ligands: Gain without pain. Trends in Endocrinology &
Metabolism, 28(4), 247–249. doi:10.1016/j.tem.2017.01.001
Google Scholar WorldCat

Rasenick, M. M., & Childers, S. R. (1989). Modification of Gs-stimulated adenylate cyclase in brain membranes by low pH
pretreatment: Correlation with altered guanine nucleotide exchange. Journal of Neurochemistry, 53, 219–225.
Google Scholar WorldCat

Reinecke, H., Weber, C., Lange, K., Simon, M., Stein, C., & Sorgatz, H. (2015). Analgesic e icacy of opioids in chronic pain: Recent
meta-analyses. British Journal of Pharmacology, 172(2), 324–333. doi:10.1111/bph.12634
Google Scholar WorldCat

Reiss, D., Ceredig, R. A., Secher, T., Boue, J., Barreau, F., Dietrich, G., & Gaveriaux-Ru , C. (2017). Mu and delta opioid receptor
knockout mice show increased colonic sensitivity. European Journal of Pain, 21(4), 623–634. doi:10.1002/ejp.965
Google Scholar WorldCat

Revici, E., Stoopen, E., Frenk, E., & Ravich, R. A. (1949). The relation of pain to local physico-chemical changes. Bulletin of the

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Institute of Applied Biology, 1, 21–38.
Google Scholar WorldCat

Richards, N., & McMahon, S. B. (2013). Targeting novel peripheral mediators for the treatment of chronic pain. British Journal of
Anaesthesiology, 111(1), 46–51. doi:10.1093/bja/aet216
Google Scholar WorldCat

Rittner, H. L., Hackel, D., Voigt, P., Mousa, S., Stolz, A., Labuz, D., … Brack, A. (2009). Mycobacteria attenuate nociceptive
responses by formyl peptide receptor triggered opioid peptide release from neutrophils. PLoS Pathogens, 5(4), e1000362.
doi:10.1371/journal.ppat.1000362
Google Scholar WorldCat

Rivat, C., & Ballantyne, J. (2016). The dark side of opioids in pain management: Basic science explains clinical observation. Pain
Reports, 1, e570.
Google Scholar WorldCat

Roberts, N. J., Vogelstein, J. T., Parmigiani, G., Kinzler, K. W., Vogelstein, B., & Velculescu, V. E. (2012). The predictive capacity of
personal genome sequencing. Science Translational Medicine, 4(133), 133ra158. doi:10.1126/scitranslmed.3003380
Google Scholar WorldCat

Roeckel, L. A., Le Coz, G. M., Gaveriaux-Ru , C., & Simonin, F. (2016). Opioid-induced hyperalgesia: Cellular and molecular
mechanisms. Neuroscience, 338, 160–182. doi:10.1016/j.neuroscience.2016.06.029
Google Scholar WorldCat

Roeckel, L.-A., Utard, V., Reiss, D., Mouheiche, J., Maurin, H., Robé, A., Audouard, E., Wood, J. N., Goumon, Y., Simonin, F., &
Gaveriaux-Ru , C. (2017). Morphine-induced hyperalgesia involves mu opioid receptors and the metabolite morphine-3-
glucuronide. Scientific Reports, 7(1), 10406.
Google Scholar WorldCat

Roques, B. P., Fournie-Zaluski, M. C., & Wurm, M. (2012). Inhibiting the breakdown of endogenous opioids and cannabinoids to
alleviate pain. Nature Reviews. Drug Discovery, 11(4), 292–310. doi:10.1038/nrd3673
Google Scholar WorldCat

Ross, J. R., Riley, J., Quigley, C., & Welsh, K. I. (2006). Clinical pharmacology and pharmacotherapy of opioid switching in cancer
patients. The Oncologist, 11(7), 765–773. doi:10.1634/theoncologist.11-7-765
Google Scholar WorldCat

p. 764 Rougeot, C., Robert, F., Menz, L., Bisson, J. F., & Messaoudi, M. (2010). Systemically active human opiorphin is a potent yet non-
addictive analgesic without drug tolerance e ects. Journal of Physiology & Pharmacology, 61(4), 483–490. Retrieved from
https://www.ncbi.nlm.nih.gov/pubmed/20814077
Google Scholar WorldCat

Rowan, M. P., Szteyn, K., Doyle, A. P., Gomez, R., Henry, M. A., & Jeske, N. A. (2014). Beta-arrestin-2-biased agonism of delta opioid
receptors sensitizes transient receptor potential vanilloid type 1 (TRPV1) in primary sensory neurons. Molecular Pain, 10, 50.
doi:10.1186/1744-8069-10-50
Google Scholar WorldCat

Rozen, D., & Grass, G. W. (2005). Perioperative and intraoperative pain and anesthetic care of the chronic pain and cancer pain
patient receiving chronic opioid therapy. Pain Practice: The O icial Journal of the World Institute of Pain, 5(1), 18–32.
doi:10.1111/j.1533-2500.2005.05104.x
Google Scholar WorldCat

Ruscheweyh, R., Wilder-Smith, O., Drdla, R., Liu, X. G., & Sandkuhler, J. (2011). Long-term potentiation in spinal nociceptive
pathways as a novel target for pain therapy. Molecular Pain, 7, 20. doi:10.1186/1744-8069-7-20
Google Scholar WorldCat

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Sasaki, K., Sumiyoshi, A., Nonaka, H., Kasahara, Y., Ikeda, K., Hall, F. S., … Sora, I. (2015). Specific regions display altered grey
matter volume in mu-opioid receptor knockout mice: MRI voxel-based morphometry. British Journal of Pharmacology, 172(2),
654–667. doi:10.1111/bph.12807
Google Scholar WorldCat

Schepers, R. J., Mahoney, J. L., Gehrke, B. J., & Shippenberg, T. S. (2008). Endogenous kappa-opioid receptor systems inhibit
hyperalgesia associated with localized peripheral inflammation. Pain, 138(2), 423–439. doi:10.1016/j.pain.2008.01.023
Google Scholar WorldCat

Schneider, J. P., & Kirsh, K. L. (2010). Defining clinical issues around tolerance, hyperalgesia, and addiction: A quantitative and
qualitative outcome study of long-term opioid dosing in a chronic pain practice. Journal of Opioid Management, 6(6), 385–395.
Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/21268999
Google Scholar WorldCat

Schreiter, A., Gore, C., Labuz, D., Fournie-Zaluski, M. C., Roques, B. P., Stein, C., & Machelska, H. (2012). Pain inhibition by blocking
leukocytic and neuronal opioid peptidases in peripheral inflamed tissue. FASEB Journal, 26(12), 5161–5171.
Google Scholar WorldCat

Schrepf, A., Harper, D. E., Harte, S. E., Wang, H., Ichesco, E., Hampson, J. P., … Harris, R. E. (2016). Endogenous opioidergic
dysregulation of pain in fibromyalgia: A PET and fMRI study. Pain, 157(10), 2217–2225. doi:10.1097/j.pain.0000000000000633
Google Scholar WorldCat

Schuchat, A., Houry, D., & Guy, G. P., Jr. (2017). New data on opioid use and prescribing in the United States. Journal of the
American Medical Association, 318(5), 425–426. doi:10.1001/jama.2017.8913
Google Scholar WorldCat

Schumacher, M. A., Basbaum, A. I., & Naidu, R. K. (2015). Opioid agonists and antagonists. In B. G. Katzung & A. J. Trevor (Eds.),
Basic and Clinical Pharmacology (13th ed., pp. 531–551). New York: McGraw-Hill Medical.
Google Scholar Google Preview WorldCat COPAC

Selley, D. E., Breivogel, C. S., & Childers, S. R. (1993). Modification of G protein-coupled functions by low-pH pretreatment of
membranes from NG108-15 cells: Increase in opioid agonist e icacy by decreased inactivation of G proteins. Molecular
Pharmacology, 44(4), 731–741. Retrieved from http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?
cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=8232223
Google Scholar WorldCat

Sharp, B. M. (2006). Multiple opioid receptors on immune cells modulate intracellular signaling. Brain, Behavior, & Immunity,
20(1), 9–14. Retrieved from http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?
cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=16364815
Google Scholar WorldCat

Silver, I. A. (1975). Measurement of pH and ionic composition of pericellular sites. Philosophical Transactions of the Royal Society
of London, B. Biological Sciences, 271(912), 261–272. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/239420
Google Scholar WorldCat

p. 765 Simmen, H. P., & Blaser, J. (1993). Analysis of pH and pO2 in abscesses, peritoneal fluid, and drainage fluid in the presence or
absence of bacterial infection during and a er abdominal surgery. American Journal of Surgery, 166(1), 24–27. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/8328625
Google Scholar WorldCat

Simonin, F., Valverde, O., Smadja, C., Slowe, S., Kitchen, I., Dierich, A., … Kie er, B. L. (1998). Disruption of the kappa-opioid
receptor gene in mice enhances sensitivity to chemical visceral pain, impairs pharmacological actions of the selective kappa-
agonist U-50,488H and attenuates morphine withdrawal. EMBO Journal, 17(4), 886–897. doi:10.1093/emboj/17.4.886
Google Scholar WorldCat

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Sirohi, S., Aldrich, J. V., & Walker, B. M. (2016). Species di erences in the e ects of the kappa-opioid receptor antagonist
zyklophin. Alcohol, 51, 43–49. doi:10.1016/j.alcohol.2015.11.012
Google Scholar WorldCat

Sitbon, P., Van Elstraete, A., Hamdi, L., Juarez-Perez, V., Mazoit, J. X., Benhamou, D., & Rougeot, C. (2016). STR-324, a stable
analog of opiorphin, causes analgesia in postoperative pain by activating endogenous opioid receptor-dependent pathways.
Anesthesiology, 125(5), 1017–1029. doi:10.1097/ALN.0000000000001320
Google Scholar WorldCat

Sitte, N., Busch, M., Mousa, S. A., Labuz, D., Rittner, H., Gore, C., … Schäfer, M. (2007). Lymphocytes upregulate signal sequence-
encoding proopiomelanocortin mRNA and beta-endorphin during painful inflammation in vivo. Journal of Neuroimmunology,
183(1–2), 133–145. Retrieved from http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?
cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=17223201
Google Scholar WorldCat

Smith, M. L., Hostetler, C. M., Heinricher, M. M., & Ryabinin, A. E. (2016). Social transfer of pain in mice. Science Advances, 2(10),
e1600855. doi:10.1126/sciadv.1600855
Google Scholar WorldCat

Smith, S. M., Dart, R. C., Katz, N. P., Paillard, F., Adams, E. H., Comer, S. D., … Dworkin, R. H.; Analgesic, Anesthetic, and Addiction
Clinical Trials, Translations, Innovations, Opportunities, and Networks (ACTTION) public-private partnership. (2013).
Classification and definition of misuse, abuse, and related events in clinical trials: ACTTION systematic review and
recommendations. Pain, 154(11), 2287–2296. doi:10.1016/j.pain.2013.05.053
Google Scholar WorldCat

Smith, S. M., Dworkin, R. H., Turk, D. C., Baron, R., Polydefkis, M., Tracey, I., … Witter, J. (2017). The potential role of sensory
testing, skin biopsy, and functional brain imaging as biomarkers in chronic pain clinical trials: IMMPACT considerations. Journal
of Pain, 18(7), 757–777. doi:10.1016/j.jpain.2017.02.429
Google Scholar WorldCat

Soergel, D. G., Subach, R. A., Burnham, N., Lark, M. W., James, I. E., Sadler, B. M., … Webster, L. R. (2014). Biased agonism of the
mu-opioid receptor by TRV130 increases analgesia and reduces on-target adverse e ects versus morphine: A randomized,
double-blind, placebo-controlled, crossover study in healthy volunteers. Pain, 155(9), 1829–1835. doi:10.1016/j.pain.2014.06.011
Google Scholar WorldCat

Sounier, R., Mas, C., Steyaert, J., Laeremans, T., Manglik, A., Huang, W., … Granier, S. (2015). Propagation of conformational
changes during mu-opioid receptor activation. Nature, 524(7565), 375–378. doi:10.1038/nature14680
Google Scholar WorldCat

Spahn, V., Del Vecchio, G., Labuz, D., Rodriguez-Gaztelumendi, A., Massaly, N., Temp, J., … Stein, C. (2017). A nontoxic pain killer
designed by modeling of pathological receptor conformations. Science, 355(6328), 966–969. doi:10.1126/science.aai8636
Google Scholar WorldCat
Spahn, V., Fischer, O., Endres-Becker, J., Schäfer, M., Stein, C., & Zöllner, C. (2013). Opioid withdrawal increases transient receptor
potential vanilloid 1 activity in a protein kinase A-dependent manner. Pain, 154 (4), 598–608. doi:10.1016/j.pain.2012.12.026
Google Scholar WorldCat

Spahn, V., & Stein, C. (2017). Targeting delta opioid receptors for pain treatment: Drugs in phase I and II clinical development.
Expert Opinion on Investigational Drugs, 26(2), 155–160. doi:10.1080/13543784.2017.1275562
Google Scholar WorldCat

p. 766 Spetea, M., Eans, S. O., Ganno, M. L., Lantero, A., Mairegger, M., Toll, L., … McLaughlin, J. P. (2017). Selective kappa receptor
partial agonist HS666 produces potent anti-nociception without inducing aversion a er i.c.v. administration in mice. British
Journal of Pharmacology, 174(15), 2444–2456. doi:10.1111/bph.13854

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Google Scholar WorldCat

Starnowska, J., Costante, R., Guillemyn, K., Popiolek-Barczyk, K., Chung, N. N., Lemieux, C., … Przewlocka, B. (2017). Analgesic
properties of opioid/NK1 multitarget ligands with distinct in vitro profiles in naïve and chronic constriction injury mice. ACS
Chemical Neuroscience, 8(10), 2315–2324. doi:10.1021/acschemneuro.7b00226
Google Scholar WorldCat

Stein, C. (1993). Peripheral mechanisms of opioid analgesia. Anesthesia & Analgesia, 76, 182–191.
Google Scholar WorldCat

Stein, C. (1995). The control of pain in peripheral tissue by opioids. New England Journal of Medicine, 332(25), 1685–1690.
Retrieved from http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=7760870
Google Scholar WorldCat

Stein, C. (1997). Opioid treatment of chronic nonmalignant pain. Anesthesia & Analgesia, 84(4), 912–914.
Google Scholar WorldCat

Stein, C. (2013a). Opioids, sensory systems and chronic pain. European Journal of Pharmacology, 716, 179–187.
doi:10.1016/j.ejphar.2013.01.076
Google Scholar WorldCat

Stein, C. (2013b). Towards safer and more e ective analgesia. Veterinary Journal, 196(1), 6–7. doi:10.1016/j.tvjl.2012.09.013
Google Scholar WorldCat

Stein, C. (2016). Opioid Receptors. Annual Review of Medicine, 67, 433–451. doi:10.1146/annurev-med-062613-093100
Google Scholar WorldCat

Stein, C. (2018). Pain. In J.-M. Cavaillon & M. Singer (Eds.), Inflammation: From Molecular and Cellular Mechanisms to the Clinic
(pp. 891–914). Weinheim, Germany: Wiley-VCH.
Google Scholar Google Preview WorldCat COPAC

Stein, C., Comisel, K., Haimerl, E., Yassouridis, A., Lehrberger, K., Herz, A., & Peter, K. (1991). Analgesic e ect of intraarticular
morphine a er arthroscopic knee surgery. New England Journal of Medicine, 325(16), 1123–1126. Retrieved from
http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=1653901
Google Scholar WorldCat

Stein, C., Hassan, A. H., Przewlocki, R., Gramsch, C., Peter, K., & Herz, A. (1990). Opioids from immunocytes interact with
receptors on sensory nerves to inhibit nociception in inflammation. Proceedings of the National Academy of Sciences USA, 87(15),
5935–5939. Retrieved from http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?
cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=1974052
Google Scholar WorldCat

Stein, C., Hassan, A. H. S., Lehrberger, K., Giefing, J., & Yassouridis, A. (1993). Local analgesic e ect of endogenous opioid
peptides. Lancet, 342, 321–324.
Google Scholar WorldCat

Stein, C., & Kopf, A. (2015). Anesthesia and treatment of chronic pain. In R. D. Miller (Ed.), Millerʼs Anesthesia, 8th Edition (Vol. 8,
pp. 1898–1918). Philadelphia, PA: Elsevier Saunders.
Google Scholar Google Preview WorldCat COPAC

Stein, C., & Küchler, S. (2013). Targeting inflammation and wound healing by opioids. Trends in Pharmacological Sciences, 34(6),
303–312. doi:10.1016/j.tips.2013.03.006
Google Scholar WorldCat

Stein, C., & Machelska, H. (2011). Modulation of peripheral sensory neurons by the immune system: Implications for pain

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


therapy. Pharmacological Reviews, 63, 860–881.
Google Scholar WorldCat

Stein, C., Pflüger, M., Yassouridis, A., Hoelzl, J., Lehrberger, K., Welte, C., & Hassan, A. H. (1996). No tolerance to peripheral
morphine analgesia in presence of opioid expression in inflamed synovia. Journal of Clinical Investigation, 98(3), 793–799.
doi:10.1172/JCI118852
Google Scholar WorldCat

Stevens, C. W. (2015). Bioinformatics and evolution of vertebrate nociceptin and opioid receptors. Vitamins & Hormones, 97, 57–
94. doi:10.1016/bs.vh.2014.10.002
Google Scholar WorldCat

p. 767 Stubbs, M., McSheehy, P. M., & Gri iths, J. R. (1999). Causes and consequences of acidic pH in tumors: A magnetic resonance
study. Advances in Enzyme Regulation, 39, 13–30. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/10470364
Google Scholar WorldCat

Szigethy, E., Knisely, M., & Drossman, D. (2017). Opioid misuse in gastroenterology and non-opioid management of abdominal
pain. Nature Reviews Gastroenterology & Hepatology, 15(3), 168–180. doi:10.1038/nrgastro.2017.141
Google Scholar WorldCat

Treuha , P. S., & McCarty, D. J. (1971). Synovial fluid pH, lactate, oxygen and carbon dioxide partial pressure in various joint
diseases. Arthritis & Rheumatism, 14(4), 475–484. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/5564921
Google Scholar WorldCat

Tulamo, R. M., Bramlage, L. R., & Gabel, A. A. (1989). Sequential clinical and synovial fluid changes associated with acute
infectious arthritis in the horse. Equine Veterinary Journal, 21(5), 325–331. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/2776718
Google Scholar WorldCat

Turnaturi, R., Arico, G., Ronsisvalle, G., Parenti, C., & Pasquinucci, L. (2016). Multitarget opioid ligands in pain relief: New players
in an old game. European Journal of Medicinal Chemistry, 108, 211–228. doi:10.1016/j.ejmech.2015.11.028
Google Scholar WorldCat

Tzschentke, T. M., Linz, K., Frosch, S., & Christoph, T. (2017). Antihyperalgesic, antiallodynic, and anti-nociceptive e ects of
cebranopadol, a novel potent nociceptin/orphanin FQ and opioid receptor agonist, a er peripheral and central administration in
rodent models of neuropathic pain. Pain Practice: The O icial Journal of the World Institute of Pain, 17(8), 1032–1041.
doi:10.1111/papr.12558
Google Scholar WorldCat

Vadivelu, N., Mitra, S., & Hines, R. L. (2011). Peripheral opioid receptor agonists for analgesia: A comprehensive review. Journal of
Opioid Management, 7(1), 55–68. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/21434585
Google Scholar WorldCat

Valdez-Morales, E., Guerrero-Alba, R., Ochoa-Cortes, F., Benson, J., Spreadbury, I., Hurlbut, D., … Vanner, S. (2013). Release of
endogenous opioids during a chronic IBD model suppresses the excitability of colonic DRG neurons. Neurogastroenterology &
Motility, 25(1), 39–46, e34. doi:10.1111/nmo.12008
Google Scholar WorldCat

Valverde, A., & Gunkel, C. I. (2005). Pain management in horses and farm animals. Journal of Veterinary Emergency & Critical
Care, 15(4), 295–307.
Google Scholar WorldCat

Varadi, A., Marrone, G. F., Palmer, T. C., Narayan, A., Szabo, M. R., Le Rouzic, V., … Majumdar, S. (2016).
Mitragynine/corynantheidine pseudoindoxyls as opioid analgesics with mu agonism and delta antagonism, which do not recruit
beta-arrestin-2. Journal of Medicinal Chemistry, 59(18), 8381–8397. doi:10.1021/acs.jmedchem.6b00748

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Google Scholar WorldCat

Vardy, E., Sassano, M. F., Rennekamp, A. J., Kroeze, W. K., Mosier, P. D., Westkaemper, R. B., … Roth, B. L. (2015). Single amino
acid variation underlies species-specific sensitivity to amphibian skin-derived opioid-like peptides. Chemistry & Biology, 22(6),
764–775. doi:10.1016/j.chembiol.2015.05.012
Google Scholar WorldCat

Vaupel, P. W., Frinak, S., & Bicher, H. I. (1981). Heterogeneous oxygen partial pressure and pH distribution in C3H mouse
mammary adenocarcinoma. Cancer Research, 41(5), 2008–2013. Retrieved from http://www.ncbi.nlm.nih.gov/pubmed/7214369
Google Scholar WorldCat

Vicente-Sanchez, A., & Pradhan, A. A. (2018). Ligand-directed signaling at the delta opioid receptor. Handbook of Experimental
Pharmacology, in press. doi:10.1007/164_2017_39
Google Scholar WorldCat

Violin, J. D., Crombie, A. L., Soergel, D. G., & Lark, M. W. (2014). Biased ligands at G-protein-coupled receptors: Promise and
progress. Trends in Pharmacological Sciences, 35(7), 308–316. doi:10.1016/j.tips.2014.04.007
Google Scholar WorldCat

p. 768 Viscusi, E. R., Webster, L., Kuss, M., Daniels, S., Bolognese, J. A., Zuckerman, S., … Skobieranda, F. (2016). A randomized, phase 2
study investigating TRV130, a biased ligand of the mu-opioid receptor, for the intravenous treatment of acute pain. Pain, 157(1),
264–272. doi:10.1097/j.pain.0000000000000363
Google Scholar WorldCat

Voegtlin, C. (1935). The influence of the parenteral administration of certain sugars on the pH of malignant tumors. National
Institutes of Health Bulletin, 164, 1–14.
Google Scholar WorldCat

Wacker, D., Stevens, R. C., & Roth, B. L. (2017). How ligands illuminate GPCR molecular pharmacology. Cell, 170(3), 414–427.
doi:10.1016/j.cell.2017.07.009
Google Scholar WorldCat

Walter, C., Doehring, A., Oertel, B. G., & Lötsch, J. (2013). Mu-opioid receptor gene variant OPRM1 118 A>G: A summary of its
molecular and clinical consequences for pain. Pharmacogenomics, 14(15), 1915–1925. doi:10.2217/pgs.13.187

Walwyn, W. M., Chen, W., Kim, H., Minasyan, A., Ennes, H. S., McRoberts, J. A., & Marvizon, J. C. (2016). Sustained suppression of
hyperalgesia during latent sensitization by mu-, delta-, and kappa-opioid receptors and alpha2A adrenergic receptors: Role of
constitutive activity. Journal of Neuroscience, 36(1), 204–221. doi:10.1523/JNEUROSCI.1751-15.2016
Google Scholar WorldCat

Wang, Y. X., Mao, X. F., Li, T. F., Gong, N., & Zhang, M. Z. (2017). Dezocine exhibits antihypersensitivity activities in neuropathy
through spinal mu-opioid receptor activation and norepinephrine reuptake inhibition. Scientific Reports, 7, 43137.
doi:10.1038/srep43137
Google Scholar WorldCat

Wasserman, R. A., Hassett, A. L., Harte, S. E., Goesling, J., Malino , H. L., Berland, D. W., … Brummett, C. M. (2015). Pressure pain
sensitivity in patients with suspected opioid-induced hyperalgesia. Regional Anesthesia & Pain Medicine, 40(6), 687–693.
doi:10.1097/AAP.0000000000000315
Google Scholar WorldCat

Wei, L. N., & Loh, H. H. (2011). Transcriptional and epigenetic regulation of opioid receptor genes: Present and future. Annual
Review of Pharmacology & Toxicology, 51, 75–97. doi:10.1146/annurev-pharmtox-010510-100605
Google Scholar WorldCat

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Weibel, R., Reiss, D., Karchewski, L., Gardon, O., Matifas, A., Filliol, D., … Gaveriaux-Ru , C. (2013). Mu opioid receptors on
primary a erent nav1.8 neurons contribute to opiate-induced analgesia: Insight from conditional knockout mice. PLoS ONE,
8(9), e74706. doi:10.1371/journal.pone.0074706
Google Scholar WorldCat

White, K. L., Robinson, J. E., Zhu, H., DiBerto, J. F., Polepally, P. R., Zjawiony, J. K., … Roth, B. L. (2015). The G protein-biased
kappa-opioid receptor agonist RB-64 is analgesic with a unique spectrum of activities in vivo. Journal of Pharmacology &
Experimental Therapeutics, 352(1), 98–109. doi:10.1124/jpet.114.216820
Google Scholar WorldCat

Williams, J. T., Ingram, S. L., Henderson, G., Chavkin, C., von Zastrow, M., Schulz, S., … Christie, M. J. (2013). Regulation of mu-
opioid receptors: Desensitization, phosphorylation, internalization, and tolerance. Pharmacological Reviews, 65(1), 223–254.
doi:10.1124/pr.112.005942
Google Scholar WorldCat

Wisner, A., Dufour, E., Messaoudi, M., Nejdi, A., Marcel, A., Ungeheuer, M. N., & Rougeot, C. (2006). Human opiorphin, a natural
anti-nociceptive modulator of opioid-dependent pathways. Proceedings of the National Academy of Sciences USA, 103(47),
17979–17984. doi:10.1073/pnas.0605865103
Google Scholar WorldCat

Woo, Y. C., Park, S. S., Subieta, A. R., & Brennan, T. J. (2004). Changes in tissue pH and temperature a er incision indicate acidosis
may contribute to postoperative pain. Anesthesiology, 101(2), 468–475. Retrieved from
http://www.ncbi.nlm.nih.gov/pubmed/15277931
Google Scholar WorldCat

Wu, H., Wacker, D., Mileni, M., Katritch, V., Han, G. W., Vardy, E., … Stevens, R. C. (2012). Structure of the human kappa-opioid
receptor in complex with JDTic. Nature, 485(7398), 327–332. doi:10.1038/nature10939
Google Scholar WorldCat

p. 769 Xu, M., Petraschka, M., McLaughlin, J. P., Westenbroek, R. E., Caron, M. G., Lefkowitz, R. J., … Chavkin, C. (2004). Neuropathic pain
activates the endogenous kappa opioid system in mouse spinal cord and induces opioid receptor tolerance. Journal of
Neuroscience, 24(19), 4576–4584. doi:10.1523/JNEUROSCI.5552-03.2004
Google Scholar WorldCat

Yadlapalli, J. S., Ford, B. M., Ketkar, A., Wan, A., Penthala, N. R., Eo , R. L., … Crooks, P. A. (2016). Antinociceptive e ects of the 6-
O-sulfate ester of morphine in normal and diabetic rats: Comparative role of mu- and delta-opioid receptors. Pharmacology
Research, 113(Pt A), 335–347. doi:10.1016/j.phrs.2016.09.012
Google Scholar WorldCat

Yaksh, T. L., Woller, S. A., Ramachandran, R., & Sorkin, L. S. (2015). The search for novel analgesics: Targets and mechanisms.
F1000Prime Reports, 7, 56. doi:10.12703/P7-56
Google Scholar WorldCat

Yekkirala, A. S., Roberson, D. P., Bean, B. P., & Woolf, C. J. (2017). Breaking barriers to novel analgesic drug development. Nature
Reviews Drug Discovery, 16(8), 545–564. doi:10.1038/nrd.2017.87
Google Scholar WorldCat

Yen, L. T., Hsieh, C. L., Hsu, H. C., & Lin, Y. W. (2017). Targeting ASIC3 for relieving mice fibromyalgia pain: Roles of
electroacupuncture, opioid, and adenosine. Scientific Reports, 7, 46663. doi:10.1038/srep46663
Google Scholar WorldCat

Yi, S. P., Kong, Q. H., Li, Y. L., Pan, C. L., Yu, J., Cui, B. Q., … Chang, K. J. (2017). The opioid receptor triple agonist DPI-125 produces
analgesia with less respiratory depression and reduced abuse liability. Acta Pharmacologica Sinica, 38(7), 977–989.
doi:10.1038/aps.2017.14
Google Scholar WorldCat

Downloaded from https://academic.oup.com/edited-volume/34282/chapter/290641250 by OUP site access user on 03 April 2024


Zeng, C., Gao, S. G., Cheng, L., Luo, W., Li, Y. S., Tu, M., … Lei, G. H. (2013). Single-dose intra-articular morphine a er arthroscopic
knee surgery: A meta-analysis of randomized placebo-controlled studies. Arthroscopy, 29(8), 1450–1458, e1452.
doi:10.1016/j.arthro.2013.04.005
Google Scholar WorldCat

Zheng, Z., Huang, X. P., Mangano, T. J., Zou, R., Chen, X., Zaidi, S. A., … Katritch, V. (2017). Structure-based discovery of new
antagonist and biased agonist chemotypes for the kappa opioid receptor. Journal of Medicinal Chemistry, 60(7), 3070–3081.
doi:10.1021/acs.jmedchem.7b00109
Google Scholar WorldCat

Zöllner, C., Mousa, S. A., Fischer, O., Rittner, H. L., Shaqura, M., Brack, A., … Schafer, M. (2008). Chronic morphine use does not
induce peripheral tolerance in a rat model of inflammatory pain. Journal of Clinical Investigation, 118(3), 1065–1073.
doi:10.1172/JCI25911
Google Scholar WorldCat

Zöllner, C., Shaqura, M. A., Bopaiah, C. P., Mousa, S. A., Stein, C., & Schäfer, M. (2003). Painful inflammation-induced increase in
mu-opioid receptor binding and G-protein coupling in primary a erent neurons. Molecular Pharmacology, 64(2), 202–210.
Google Scholar WorldCat

You might also like