Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

CNS Drugs (2019) 33:943–955

https://doi.org/10.1007/s40263-019-00660-0

CURRENT OPINION

Opioid‑Induced Tolerance and Hyperalgesia


Sebastiano Mercadante1,2 · Edoardo Arcuri3 · Angela Santoni4

Published online: 1 October 2019


© Springer Nature Switzerland AG 2019

Abstract
Opioids are very potent and efficacious drugs, traditionally used for both acute and chronic pain conditions. However, the use
of opioids is frequently associated with the occurrence of adverse effects or clinical problems. Other than adverse effects and
dependence, the development of tolerance is a significant problem, as it requires increased opioid drug doses to achieve the
same effect. Mechanisms of opioid tolerance include drug-induced adaptations or allostatic changes at the cellular, circuitry,
and system levels. Dose escalation in long-term opioid therapy might cause opioid-induced hyperalgesia (OIH), which is a
state of hypersensitivity to painful stimuli associated with opioid therapy, resulting in exacerbation of pain sensation rather
than relief of pain. Various strategies may provide extra-opioid analgesia. There are drugs that may produce independ-
ent analgesic effects. A tailored treatment provided by skilled personnel, in accordance with the individual condition, is
mandatory. Any treatment aimed at reducing opioid consumption may be indicated in these circumstances. Interventional
techniques able to decrease the pain input may allow a decrease in the opioid dose, thus reverting the mechanisms produc-
ing tolerance of OIH. Intrathecal therapy with local anesthetics and a sympathetic block are the most common techniques
utilized in these circumstances.

1 Introduction
Key Points
Chronic pain is a major health issue that impacts quality of
The development of tolerance is the main problem dur- life. About one-third of the population in western countries
ing opioid treatment as it requires increased opioid drug experiences chronic pain, with the cost related to this esti-
doses to achieve the previous level of analgesia. mated to be about $US600 billion annually [1, 2]. Despite
Mechanisms of opioid tolerance include drug-induced the development of new analgesic substances for treating
adaptations or allostatic changes at the cellular, circuitry, pain, opioids still remain the most frequent class of analge-
and system levels. sics used in various clinical settings. Opioids are very potent
and efficacious drugs, traditionally used for both acute and
Dose escalation in long-term opioid therapy might result chronic pain conditions. When opioids are used for chronic
in a state of hypersensitivity to painful stimuli, known as pain, they are administered for a prolonged time period.
opioid-induced hyperalgesia (OIH), resulting in exacer- However, the use of opioids is frequently associated with
bation of pain rather than pain relief. the occurrence of adverse effects or clinical problems.
Possible strategies to mitigate, reverse, or prevent opioid- Long-term administration of morphine and other opioids
induced tolerance and OIH are the use of adjuvant can lead to the development of analgesic tolerance. Toler-
analgesics or opioid switching. ance has been defined as a reduction in effect following
prolonged drug administration that results in a decrease of

2
* Sebastiano Mercadante Palliative/Supportive Care and Rehabilitation, MD Anderson,
terapiadeldolore@lamaddalenanet.it Houston, TX, USA
3
Edoardo Arcuri Regina Elena Cancer Institute, Rome, Italy
edoardo.arcuri@fastwebnet.it 4
University of Rome, Rome, Italy
Angela Santoni
angela.santoni@uniroma1.it
1
Main Regional Center of Supportive/Palliative Care, La
Maddalena Cancer Center, Palermo, Italy

Vol.:(0123456789)
944 S. Mercadante et al.

drug potency, characterized by a shift to the right in the Another problem with tolerance is that the management of
dose–response curve. The term tolerance itself has been pain in opioid addicts is extremely challenging, as metha-
used to describe multiple processes. While associative tol- done-maintained patients are more refractory to the analge-
erance is related to conditioning, pharmacological tolerance sic effects of opioids just because of the high level of toler-
is related to specific drug actions. Pharmacological tolerance ance [8].
may be dispositional, due to pharmacokinetic changes or Opioids generate opponent processes in both nociceptive
pharmacodynamics. The latter is more common and char- circuits and circuits modulating mood. The involvement of
acterized by a decline in analgesia related to complex pro- different circuits contributes to the production of an addicted
cesses of neural adaptation. There is a large intra-individual state favoring the hedonic aspects of drug taking. Among
and inter-individual variability in the development of toler- these adverse effects, prolonged opioid therapy is associated
ance. This phenomenon develops at varying rates and can with tolerance and OIH, which are relevant for the inevita-
occur both after short-term dosing or accrue more gradually ble consequences on the clinical responses. While the pro-
with long-term dosing. Tolerance to adverse effects has its gressive decrease in analgesia produced by an opioid given
own favorable aspect as it allows the therapeutic window to long-term may be compensated for with an increased dose to
be enlarged. From a clinical perspective, the development achieve the same level of the previous analgesia, with OIH
of tolerance may become the ‘driving force’ for dose esca- there is a development of hypersensitivity to painful stimuli
lation, which can be constant during long-term administra- associated with opioid therapy, resulting in exacerbation of
tion and can have an accelerated phase at the beginning of pain sensation rather than relief from pain.
the treatment as well as at other times during the course It is relevant to distinguish between tolerance and OIH
of treatment. Tolerance may be a major contributing factor [3]. Tolerance is exhibited with most opioid effects but not
in declining opioid effects, and may create a situation of to miosis and constipation. The increase in doses overcomes
poor responsiveness if adverse effects occur as the dose is this problem and the patient will be relieved of his pain. In
increased [3]. OIH there is a state of nociceptive sensitization induced by
There are some factors that influence the development of an acute as well as a prolonged exposure to opioids. The
tolerance, including the interactions between the drug and level of pain experienced might be the same or often might
the opioid receptors, doses, and frequency of administra- be different. However, it cannot be resolved by increasing
tion. Many mechanisms contribute to opioid tolerance at a the dose of the drug, which instead can aggravate the pain.
behavioral level. These processes include the upregulation of This phenomenon can be relieved only by decreasing the
drug metabolism (i.e., metabolic tolerance), desensitization dose of opioid or discontinuing it while using other strate-
of receptor signaling, downregulation of receptors, and com- gies to compensate the underlying pain condition [9]. This
pensatory/opponent processes [4]. From a clinical perspec- clinical eventuality is quite complex, as a negative feedback
tive, analgesic tolerance is noteworthy, as the management loop can be elicited so that the development of tolerance
of such patients during acute episodes of care is a challenge often induces physicians to increase opioid doses. This
because they will have a significantly longer length of hos- decision, in turn, facilitates the development of OIH. The
pital stay, are more likely to be readmitted within 30 days, clinical implications are that dose escalation in long-term
and have a higher risk of mortality and co-morbidities [5]. opioid therapy might cause OIH, producing a vicious cycle
Indeed, the progressive increase of opioid doses can induce of opioid escalation and anxiety both for the physician and
or exacerbate undiagnosed opioid-induced hyperalgesia patient [10].
(OIH), which in turn increases the occurrence of behavioral Like methadone- and buprenorphine-maintained patients,
analgesic tolerance and is often misdiagnosed as a progres- heroin-dependent patients have been shown to be signifi-
sion of disease-producing pain. cantly hyperalgesic to cold-pressor pain in comparisons
Tolerance is a relevant component of dependence and with non-drug users [11]. It has been reported that pain-
addiction liability. The US Food and Drug Administration free injury sites can temporarily become painful again when
(FDA) defines tolerance as an opioid dose of 60 morphine stopping opioids, a phenomenon that has been recently
milligram equivalents daily [6]. Indeed, tolerance to opioid- named withdrawal-associated injury-site pain [12]. Finally,
induced respiratory depression is the other face of the coin, mini-opioid withdrawal, often associated with inappropriate
because opioid tolerance can result in protection against a treatments, has been associated with OIH [13].
fatal complication with increasing doses. However, it can Acute opioid tolerance, defined as an increase in the
also cause an increase in mortality when addicted patients required opioid dose to maintain adequate analgesia, and
take the same dose of opioid in a different environment/ OIH, defined as a decreased pain threshold after long-
context [7] or when they relapse after a period of abstinence. term opioid treatment, should be suspected with any unex-
In these patients, in fact, the level of tolerance abates and plained pain report unassociated with disease progres-
an opioid dose may induce life-threatening consequences. sion. In particular, exposure to short-acting opioids, such as
Opioid-Induced Tolerance and Hyperalgesia 945

remifentanil, seems more likely to be responsible for post- In the PAG–RVM connection, opioid tolerance is associated
operative high pain scores, high morphine consumption, and with changes in a number of signaling cascades.
hypersensitivity to pain. In contrast to the acute inhibitory effect of opioids on
In this review the mechanisms of tolerance, including cyclic adenosine monophosphate (cAMP) production, long-
drug-induced adaptations or allostatic changes at the cellu- term treatment upregulates cAMP. Direct activation of the
lar, circuitry, and system levels, that require increased opioid G-protein coupled opioid receptor induces the dissociation
drug doses to achieve the same effect, as well the progres- of Gα and Gβγ subunits. The consequence of the dissociated
sion to the state of hyper-excitation present with OIH are G-protein subunits is the inhibition of VGCCs and activation
described. of inward-rectifying potassium channels. In addition, down-
stream adenylate cyclase enzymes are inhibited, decreas-
ing cAMP levels and thus, the activation of the adenylyl
2 Mechanism of Tolerance cyclase–cAMP–protein kinase A (PKA) pathway, which
and Opioid‑Induced Hyperalgesia results in a hyperpolarization and decreased neurotransmit-
ter release in the PAG, corresponding to anti-nociception
2.1 The Descending System [22, 23]. Opioid binding also stimulates receptor phospho-
rylation by the G-protein–coupled receptor (GPCR) kinase
The midbrain periaqueductal grey (PAG) is a key site of the (GRK), which leads to β-arrestin recruitment to the receptor.
analgesic activity of opioids within the central nervous sys- This results in receptor desensitization and internalization
tem. Several studies have identified the postsynaptic adap- (Fig. 1, left panel).
tations induced by long-term opioid administration in PAG Long-term use of morphine produces adaptations that
and other brain regions [14, 15]. Opioids activate an anal- contribute to opioid tolerance within all of these down-
gesic system within the PAG, disinhibiting γ-aminobutyric stream signaling pathways. The sustained β-arrestin bind-
acid (GABA)ergic activity by reducing the GABAergic inhi- ing to the receptor leads to internalization, degradation, and
bition [16]. The GABAergic disinhibition is produced by a decreased number of membrane receptors (Fig. 1, right
two independent mechanisms within the PAG: one is a direct panel). Increased adenylate cyclase activity and PKA activa-
postsynaptic inhibition of GABAergic neurons; the second is tion, activation of N-methyl-d-aspartate (NMDA) receptors,
a presynaptic inhibition of release of GABA from nerve ter- and downregulation of glutamate receptors are all implicated
minals [16–18]. These mechanisms facilitate the activation in attenuated analgesic effects, aggravated pain intensity,
of the PAG descending analgesic system that projects via the increased tolerance, and OIH. In this regard, evidence is
medulla to the dorsal horn. The postsynaptic mechanisms available on the ability of NMDA receptor antagonists to
have been focused on in different studies showing that long- limit the development of opioid tolerance [24, 25].
term opioid administration induces a reduction in postsyn- The upregulation in cAMP seems to be caused by com-
aptic coupling of μ-opioid receptors to G-protein inwardly pensatory changes in intracellular signaling, or an uncou-
rectifying ­K+ (GIRK) conductance and voltage-gated cal- pling of Gi-/Go-proteins from the receptor and a switch to
cium channels (VGCCs) within the PAG [19]. Moreover, coupling with Gs-proteins [26]. The mechanisms underlying
long-term opioid administration also reduces presynaptic receptor downregulation and tolerance may depend on the
μ-opioid receptor inhibition by decreasing the size of the specific agonist. Morphine does not readily recruit β-arrestin
vesicles available for action potential dependent release or internalize the receptor, in contrast to high-efficacy ago-
[14]. This depletion leads to a reduction in μ-opioid receptor nists such as fentanyl. Such differences in signaling suggest
presynaptic inhibition of GABAergic synaptic transmission. that the mechanisms can be different. Morphine could use a
Thus, long-term opioid administration reduces the presyn- G-protein-dependent mechanism, while other opioids may
aptic opioid inhibition within GABAergic nerve terminals. use a β-arrestin-dependent mechanism [27–30].
OIH is the increased sensitivity to pain following long- Opioid tolerance can be regulated by neuropeptides
term opioid treatment. Hyperalgesia may manifest as opi- within the descending pain pathway. Cholecystokinin (CCK)
oid-induced tolerance since increased sensitivity to pain within the PAG-RVM-dorsal horn pathway regulates mor-
would counteract the pain-relieving effects of opioids. The phine tolerance [31, 32], as demonstrated by morphine toler-
increased activation of the descending pain pathway by opi- ance being blocked with CCK receptor antagonists injected
oids produces neuroadaptations within the rostroventral- into the PAG [31]. Injection of CCK in the RVM blocks opi-
medial medulla (RVM) that result in hyperalgesia [20]. oid activation of off-cells, which activate descending anti-
Long-term administration of opioids produces an increase nociception, thus reducing the analgesic effects of morphine
in the number of active on-cells, which lowers the threshold [31, 33].
to noxious stimuli [21], believed to be responsible for OIH.
946 S. Mercadante et al.

Acute opioid treatment Chronic opioid treatment

Opioid
Opioid Desensized
receptor Ca2+ opioid receptor

P P
Gα Gβ K+ GRKP P GRKP P

Adenylate
G protein β-arresn β-arresn
cyclase Recycling
Acvaon

Reduced
Internalizaon
effect
P
Receptor P
degradaon
Downstream Reduced
signaling downstream
signaling
Increased adenylate
cyclate levels,PKC
P PKA, and NMDA
P

Analgesic Tolerance and


effects insufficient analgesia Hyperalgesia

Fig. 1  Opioid receptor signaling and fate in response to acute and kinase (GRK), which leads to β-arrestin recruitment to the receptor.
chronic treatment. Direct activation of the G-protein coupled opioid This results in receptor desensitization and internalization. Long-
receptor induces the dissociation of Gα and Gβγ subunits. The conse- term morphine administration produces adaptations that contribute
quence of the dissociated G-protein subunits is the inhibition of volt- to opioid-tolerance within all these downstream signaling pathways.
age-gated calcium channels and activation of inward-rectifying potas- The sustained β-arrestin binding to the receptor leads to internali-
sium channels. Downstream adenylate cyclase enzymes are inhibited, zation, degradation, and a decreased membrane receptor number.
decreasing cyclic adenosine monophosphate levels and, thus, the Increased adenylate cyclase activity and PKA activation, activation
activation of the AC-cyclic adenosine monophosphate (cAMP)- of N-methyl-D-aspartate (NMDA) receptors, and downregulation of
protein kinase A (PKA) pathway. This results in a hyperpolarization glutamate receptors are all implicated in attenuated analgesic effects,
and decreased neurotransmitter release in the periaqueductal grey aggravated pain intensity, increased tolerance, and opioid-induced
(PAG), corresponding to anti-nociception. Opioid binding also stim- hyperalgesia. PKC protein kinase C
ulates receptor phosphorylation by the G-protein-coupled receptor

2.2 Inflammation septic shock [36]. TLR-4 is expressed on microglia, and to


a lesser degree on astrocytes, and has been implicated in
Increasing evidence indicates that the inflammatory response the development of opioid tolerance, allodynia, and OIH
is directly involved in opioid-induced tolerance and hyper- [37–41]. In addition, in the spinal cord, TLR-4 is upregu-
algesia. Opioids have been shown to trigger innate immune lated after opioid treatment [42] and its upregulation is asso-
sensors on cells present at the neuroimmune interface, lead- ciated with enhanced nociception in diverse pain models
ing to the production/release of numerous pain mediators, [41].
including cytokines, chemokines, prostaglandins, and reac- Interaction of opioids with the TLR4 accessory molecule
tive oxygen species. This finally results in sensitization and MD2 initiates a cascade of intracellular events, including
threshold lowering of neuronal firing. The repeated adminis- activation of the acid sphingomyelinase with the generation
tration of opioids activates the glia within the PAG and spi- of ceramide (Fig. 2). Ceramide is crucial for the formation of
nal cord of the descending pain pathway, initiating a cascade lipid rafts, molecular platforms that promote the assembly of
of signaling events [34, 36]. TLR4 receptor complex and receptor dimerization. Moreo-
Of particular importance in the activation of the opioid- ver, ceramide activates mitogen-activated protein kinase
induced inflammatory response is Toll-like receptor (TLR)- (MAPK) pathways and a number of transcription factors,
4, the receptor that recognizes endotoxin and generates leading to the upregulation of proinflammatory cytokine
Opioid-Induced Tolerance and Hyperalgesia 947

LPS MD2

LPS binding protein LPS


CD14

Acid
sphingomyelinase
T T
I R
M R A T
y A M R
Ceramide D P
I
8
TLR4 lipid 8 F
NADPH Ra complex
oxidase
IRAK4
IRAK1
TRAF6 RIP2
TAK1
PI3K

IKKs MAPKs
Akt

Cell survival/death
NF-κB ERK1/2 JNK p38 Molity

Inflammatory genes

Fig. 2  Toll-like receptor (TLR)-4 signaling cascade in response to (PI3K)/Akt pathway involved in the control of cell motility, sur-
lipopolysaccharide (LPS) or opioid interaction. TLR signal transduc- vival, and apoptosis; p38 and extracellular signal-regulated kinase
tion initially involves MyD88, TRAM, TIRAP, and/or TRIF adap- (ERK) 1/2 mitogen-activated protein kinase (MAPK) and nuclear
tor proteins and the interleukin-1-receptor associated kinase (IRAK) factor (NF)-κB pathways regulating the expression of genes encod-
kinases. Interaction of opioids with the TLR4 accessory molecule ing proinflammatory cytokines and chemokines; inducible nitric
MD2 initiates a cascade of intracellular events including the activa- oxide synthase (iNOS) and NADPH oxidase 2 (NOX2) leading to
tion of the acid sphingomyelinase with generation of ceramide that generation of reactive nitrogen and oxygen species, respectively; and
is crucial for the assembly of TLR4 receptor complex and receptor cyclo-oxygenase (COX)-2 involved in prostaglandin synthesis. IKK
dimerization in the lipid rafts. In addition, ceramide activates nico- IκB kinase complex, JNK c-JUN N-terminal kinase, RIP2 receptor-
tinamide adenine dinucleotide phosphate (NADPH) oxidase. TLR4 interacting protein 2, TAK1 transforming growth factor-β-activated
opioid engagement also activates the phosphatidylinositol 3-kinase kinase-1, TRAF6 tumor necrosis factor receptor-associated factor 6

expression and cyclo-oxygenase (COX)-2 with increased Of interest, ceramide is also the precursor of sphingosine-
formation of prostaglandin E2 (PGE2), and enhanced gener- 1-phosphate (S1P) receptor 1 (SIP1), which binds to specific
ation of reactive oxygen and nitrogen species and peroxyni- GPCRs expressed on neurons, astrocytes, and microglia in
trite. Spinal peroxynitrite has been reported to be involved the spinal cord and dorsal root ganglion (DRG), thus sug-
in OIH in that SRI-110, which is a superoxide-sparing per- gesting an emerging role of ceramide–S1P pathway in pain
oxynitrite decomposition catalyst, can prevent both analge- [44]. Finally, ceramide may also be involved in apoptotic
sic tolerance and OIH by lowering the morphine-induced neuronal cell death [45]. OIH has been correlated with cera-
upregulation of the proinflammatory cytokines (tumor mide upregulation in astrocytes and microglia and is pre-
necrosis factor [TNF]-α, interleukin [IL]-1β, and IL-6) and vented by administration of ceramide inhibitors [46].
increasing that of the anti-inflammatory cytokines (IL-4 and TLR4 opioid engagement also activates the phosphati-
IL-10) [43]. Thus, removal of superoxides and peroxynitrites dylinositol 3-kinase (PI3K)/AKT pathway involved in
by restoring spinal mitochondrial function may increase the the control of cell motility, survival, and apoptosis, and
levels of anti-inflammatory mediators, improving opioid the p38, extracellular signal-regulated kinase (ERK) 1/2
performance MAPK, and nuclear factor (NF)-κB pathways, regulating
948 S. Mercadante et al.

the expression of genes encoding proinflammatory cytokines 2.2.1 Inflammatory Cytokines


and chemokines as well as of nitric oxide synthase (NOS)
and nicotinamide adenine dinucleotide phosphate oxidase Among the inflammatory cytokines, a pivotal role in opioid-
(NOX)2, thus leading to generation of reactive nitrogen and induced neuroinflammation and development of tolerance is
oxygen species, respectively [47], and being COX-2 involved played by TNF-α, which initiates the cascade of inflamma-
in prostaglandin synthesis [35, 48, 49]. tory cytokines, i.e., IL-6 and IL-1β. Intrathecal administra-
Inhibition of TLR4-induced microglial activation and cer- tion of morphine results in increased TNF-α expression in
amide biosynthesis attenuates the development of opioid tol- the spinal cord, and TNF levels further increase upon long-
erance, thus suggesting a relevant role of this pathway in the term opioid treatment. Inhibition of spinal TNF receptor
development of opioid tolerance [37]. Moreover, as TLR4 signaling blocks the release of IL-6 and IL-1β, and treatment
is directly activated by opioids, it is conceivable that opioid with immunomodulatory drugs such as ibudilast (AV411)
antagonists can be used to block TLR4-mediated microglial and minocycline, which can impair, block, and even reverse
activation and production of inflammatory mediators [50]. opioid tolerance, results in decreased TNF expression levels
The mechanisms underlying the alterations of neu- [53].
ronal signaling involve glial inflammatory cytokines, There is also evidence that a combination of IL-1, TNF-
which increase the number and activate neuronal AMPA α, and IL-6 blockers [40] prevent OIH as well as the IL-1
(α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor antagonist IL-1ra [54, 55] and the anti-inflamma-
and NMDA receptors and downregulate the GABA receptors tory cytokine IL-10 [54]. Finally, TNF-α and IL-1β were
as well as the astrocyte glutamate transporter 1 (GLT-1) and found to activate high-frequency stimulation-induced hyper-
L-glutamate/L-aspartate transporter that are considered to algesia and synaptic long-term potentiation (LTP) by the
be responsible for the synaptic glutamate uptake, as well as involvement of their receptors on glial cells [56].
the outward potassium currents [51]. All these mechanisms Together, these studies indicate a clear link between
result in a global increase in neuronal excitability, reduc- cytokines, tolerance, and OIH.
ing the opioid-induced hyperpolarization of µ-containing
GABAergic neurons [35, 52].

Fig. 3  Functional interaction Chemokine


between chemokine receptors
and opioid receptors. Acute Chemokine Opioid Opioid
(short-term) and chronic (long- receptor receptor
term) opioid treatment may
involve chemokine signaling
pathways. Chemokine bind-
ing to its receptors results in
heterologous desensitization of
opioid receptors by Src family
kinase (SFK) activation, leading Acute opioid Chronic opioid
to the loss of opioid-induced
analgesia. Long-term opioid
treatment induces extracellular SFK pathway ERK pathways
signal-regulated kinase (ERK)
activation, which decreases
the nociceptive threshold and Heterologous Decreased
results in morphine-induced desensizaon
hyperalgesia and apparent loss
nocicepve threshold
of opioid-induced analgesia.
SFK activation may also cause
pain hypersensitivity through Desensizaon of Opioid-induced
a direct link with the ERK opioid receptors hyperalgesia
pathway

Loss of opioid-
induced analgesia
Opioid-Induced Tolerance and Hyperalgesia 949

2.2.2 Chemokines orphanin FQ (OFQ)/nociceptin (NOP) as well as dynor-


phin [72–74] display pro-nociceptive activities that oppose
Chemokines, which are mainly implicated in the control of the anti-nociceptive action of opioids. The pharmacologi-
leukocyte trafficking, are also involved in OIH and tolerance cal blockade of CCK2 receptors in the RVM results in a
[57]. Functional interactions between chemokine receptors blockage of the development of OIH [31]. In addition,
and opioid receptors as well as their role in pain modula- CCK receptor antagonists prevent the development of anti-
tion have been described [57] (Fig. 3). Opioid receptors and nociceptive tolerance to morphine [75].
chemokine receptors are often present together in the same Overall, these findings indicate that CCK activity in the
microenvironments and co-localize. RVM may decrease the spinal analgesic effect of opioids
In the process of pain enhancement, chemokines are by favoring the descending pain facilitatory mechanism,
secreted by the primary afferent terminal, where they act in which results in an exacerbation of spinal nociceptive
an autocrine and paracrine manner to induce the release of sensitivity.
calcitonin gene-related peptide (CGRP), substance P (SP) The activities of NPFF and OFQ/NOP are ambiguous.
and glutamate. The released chemokines also participate They induce OIH-reversing morphine analgesia following
in the sensitization of second-order neurons in the dorsal intracerebroventricular administration, while producing an
spinal cord. anti-nociceptive effect intrathecally [70, 76]. Indeed, sys-
Because of the heterologous desensitization, a num- temic administration of the NPFF receptor antagonist RF9
ber of chemokines have been reported to desensitize the prevents the development of OIH and tolerance [76, 77].
µ-opioid receptor [58]. Morphine analgesia has been shown Moreover, systemic administration of a selective antagonist
to decrease in response to chemokine infusion or chemokine (J-113397) or the genetic deletion of NOP receptor or OFQ/
receptor engagement [59], whereas it can be improved by NOP precursor genes determines an increase of nocicep-
treatment with anti-chemokine receptor-neutralizing anti- tive behavior in inflammatory pain models. Thus, the spinal
body or chemokine receptor antagonists [60]. In accordance anti-nociceptive activity may prevail over supraspinal pro-
with this, the development of analgesic tolerance or OIH nociceptive effects [78, 79].
[54] can be limited by treatment with antibodies directed A role in pathological pain is also described for the neu-
against chemokines or chemokine receptors [60, 61]. ropeptides belonging to the tachykinin family [80]. The
tachykinin neurokinin (NK)1 receptor in the spinal cord
2.3 Purinergic Signaling has been reported to be involved in glia activation during
OIH. In addition, glial cell activation induced by morphine
Several findings indicate that purinergic signaling can also withdrawal and the consequent OIH are attenuated by the
contribute to opioid tolerance. Morphine engagement of tachykinin NK1 receptor antagonist L-732,138. Indeed,
the µ receptor results in upregulation of P2X4 receptor a bivalent opioid agonist–NK1 antagonist prevented OIH
(P2X4R) and P2X7 receptor (P2X7R) on microglia [62, while not activating spinal glial cells. This observation sug-
63], and genetic and pharmacological blockade of these gests that glia activation triggered by SP and OIH occurring
receptors attenuates tolerance [63–65]. Moreover, P2X4R after prolonged morphine administration could be prevented
stimulated microglial release of the brain-derived neuro- by concomitantly targeting opioid and NK1 receptors [81].
trophic factor (BDNF) [66], which induces disinhibition of
second-order nociceptive projection neurons in the spinal 2.5 Epigenetics
dorsal horn [67, 68].
The persistent nature of pain suggests that the epigenetic
machinery may be a critical factor driving chronic pain.
2.4 Neuropeptides Indeed, epigenetic processes, comprising DNA methyla-
tion, chromatin remodeling, and non-coding RNA, have
Several neuropeptides have been shown to display pro- been increasingly implicated in the development of OIH,
nociceptive activity that opposes anti-nociceptive action dependence, and tolerance.
of opioids. They have been proposed as part of a homeo- Thus, administration of a histone acetyltransferase inhibi-
static equilibrium in which exogenous administration of tor with morphine significantly reduced development of
opioids triggers the release of these peptides, which in turn opioid-induced mechanical allodynia, thermal hyperalge-
counteract the analgesic action of opioids, thus leading to sia, tolerance, and physical dependence. Conversely, the
the development of hyperalgesia and analgesic tolerance histone deacetylase inhibitor suberoylanilide hydroxamic
[69, 70]. acid (SAHA) enhanced these responses, suggesting that the
In this regard, CCK, which is considered to be an balance of histone acetyltransferase versus histone deacety-
endogenous anti-opioid [71], neuropeptide FF (NPFF) and lase might regulate these morphine-induced changes [82].
950 S. Mercadante et al.

In addition, DNA methylation is involved in the epi- withdrawal symptoms, particularly if discontinuation is
genetic regulation of pain. Opioid users exhibit increased too rapid. OIH may disappear only when a critical level of
methylation of the opioid receptor promoter gene, thus opioid dose is reached. At the same time the previous pain
decreasing its transcription [83]. Moreover, opioids may condition may still remain untreated and efforts should be
inhibit the gene demethylation of opioid receptors [84], made to provide analgesia [90].
and the epigenetic regulation of the µ-opioid receptor gene Various strategies may be helpful in this critical condi-
(OPRM1) has been also found to contribute to opioid toler- tion, such as providing extra-opioid analgesia using drugs
ance in cancer patients [85]. that may produce independent analgesic effects. However,
Conversely, demethylation regulation has been implicated given the extreme conditions of high levels of tolerance or
in the increased expression of the BDNF in DRG neurons OIH, it is difficult to find specific evidence regarding this and
following repeated administration of morphine leading to no controlled study has ever assessed specific treatments.
OIH [86]. Thus, a tailored treatment provided by skilled personnel,
Recent evidence also indicates a role for microRNAs in accordance with the individual condition, is mandatory.
(miRs) in regulating the actions of opioid drugs through the Among the mechanisms involving tolerance and OIH,
opioid receptors. Thus, the let-7 family of miRs has been it has been reported, as noted earlier, that the descending
found to be a critical regulator of receptor function in opioid system is of paramount importance. Drugs that act by rein-
tolerance [87, 88]. A potential link between receptor expres- forcing the inhibitory descending system may be beneficial
sion and morphine treatment at the post-transcriptional level in such a context. Other than improving depression and
in which miRNA23b is involved has been reported [89]. enhancing sleep, antidepressants may provide decreases in
Thus, there are complex mechanisms able to induce perception of pain. These analgesic effects of antidepressant
tolerance and OIH. However, the interaction of these drugs are not directly related to their antidepressant activity.
mechanisms, including the descending system, inflamma- Their efficacy has been established in many painful condi-
tion, purinergic signaling, neuropeptides, and epigenetics, tions and antidepressant drugs are commonly given to cancer
remains to be ascertained. patients, particularly in the presence of a prevalent neuro-
pathic pain component [91]. However, this group of drugs
results in some common adverse effects, including antimus-
3 Therapeutic Strategies carinic effects, such as dry mouth, impaired visual accom-
modation, urinary retention, and constipation, antihistaminic
A major dilemma is faced by clinicians in differentiating effects, such as drowsiness, and sympatholytic effects, such
OIH from tolerance. This differential diagnosis is challeng- as orthostatic hypotension. Alternative tricyclics with a
ing but necessary, as the treatment may be different. Making more noradrenergic activity, such as duloxetine, are often
this even more complicated is the need to distinguish among preferred in patients predisposed to sedative, anticholinergic,
OIH, progression of the disease, clinical exacerbation of pre- or hypotensive effects.
existing pain, and the myriad of social and psychological Sodium channel-blocking agents may act both centrally
factors implicated in pain expression. From a clinical per- and peripherally to reduce ectopic impulse generation and
spective, OIH is characterized by a diffuse and less definite the activity of hyperactive wide dynamic range neurons
pain, often extending to other areas of distribution than the in the dorsal horn. It has been suggested that changes
pre-existing pain. OIH exacerbates a pre-existing painful in ion channel activity are determinant in the molecular
condition above previous pain levels. When a pre-existing mechanism of neuropathic pain. Systemic local anesthet-
pain is undertreated or, in the case of tolerance, an increase ics, carbamazepine, phenytoin, and open sodium channel
in the opioid dose is prescribed, this generally produces a blockers may be useful in states of neuronal excitation.
reduction of pain. Opioid tolerance can be overcome with These agents principally act through an inhibition of
a dose increase of the opioid previously used. Generally, a sodium channel activity in hyperactive and depolarized
30–50% dose increase produces the same level of previous nerves without interfering with normal sensory function.
analgesia. With OIH, pain often worsens with an increase This group of drugs also have some activity on presynaptic
in opioid dose. Rapid opioid escalation without reporting calcium channels and postsynaptic receptors, and conse-
any benefit should be suggestive of the development of OIH. quently on the opioid system [92].
Thus, when the intervals between these dose increases Anticonvulsant agents, such as gabapentin, pregabalin,
shorten or dose escalation becomes more rapid, it is likely carbamazepine, phenytoin, valproate, and clonazepam, have
that the opioid is going to lose its efficacy or is produc- been reported to relieve pain in peripheral and central neuro-
ing a state of excitation. In these cases, the patient has to pathic pain conditions, although contradictory results have
be weaned off the high dose of opioids. This decrease in been reported. They also inhibit NMDA receptors and also
opioid dose may lead to a transient increase in pain or mild possess other activities, including sodium channel blockade
Opioid-Induced Tolerance and Hyperalgesia 951

for some. Evidence that gabapentin is analgesic is strong of ketamine and midazolam at low doses may reverse an
and, combined with a favorable safety profile, has encour- unfavorable opioid response, reduce the level of tolerance,
aged widespread use of this drug [91], although the real and assist the opioid switching [97].
advantages in extreme conditions of OIH have never been Several studies have reported that some opioids tend to
investigated. produce less tolerance, and possibly OIH, than others. This
Anti-inflammatory drugs, both steroidal and non-steroi- property probably relies on different receptor activities,
dal, may occasionally provide additional analgesia on top of exerted, for example, by lipophilic drugs such as fentanyl,
opioid analgesia with different mechanisms and may avoid methadone, and buprenorphine, in comparison with mor-
opioid dose escalation. These drugs could reduce cancer phine [98–101].
pain by decreasing peritumoral edema or inhibiting the pro- This is the basis for the concept of opioid switching. In
duction and activity of many mediators of peripheral inflam- fact, the most well-known method to counteract the devel-
mation [93]. opment of tolerance or OIH is the discontinuation of the
α2-Adrenoceptor agonists are analgesic drugs that can offending opioid whose use had been the origin of these
synergize when coadministered with opioids. These supra- molecular processes [102]. As patients need analgesics for
additive interactions are potentially clinically beneficial. their pain, another opioid may provide a better analgesia
The spinal cord is an important site of action of synergistic with a lower intensity of adverse effects. In this sense, hyper-
interactions. Synergistic interactions between opioid and α2- algesia can be considered a sign of neurotoxicity, which is
adrenoceptor agonists could be mediated by either intercel- an adverse effect. In the clinical setting, sequential thera-
lular or intracellular mechanisms, depending on the location peutic trials with different opioids have claimed to improve
of the receptors involved. In addition, α2A-adrenoceptors are the balance between analgesia and adverse effects in condi-
also expressed on descending noradrenergic axon termi- tions where this balance is unfavourable [103]. The substi-
nals, where they act as autoreceptors to inhibit noradrena- tution of an opioid for a previous one in order to improve
line release. Opioid receptors are located in both primary the clinical response has largely been reported as opioid
afferent neurons and spinal cord neurons receiving input switching. Opioid switching has been reported to produce
from primary afferents. The α2C-adrenoceptors are known a clinical improvement in a large number of patients with
to be located primarily on spinal cord interneurons and to chronic cancer pain presenting a poor response to one opioid
a much lesser degree on incoming primary afferents. The [104–106]. This observation, which implies the presence of
synergistic interaction would be due to coincident inhibi- incomplete cross-tolerance among opioids, has contributed
tion of two neurons in series in the same anatomical path- to the conclusion that the µ-opioid analgesics differ from one
way or via a retrograde feedback mechanism. The activa- another, contradicting the concept of a uniform category of
tion of both μ- and δ-opioid receptors can produce synergy µ-receptor opioid agonists similar to morphine in respect to
with α2-adrenoceptor agonists [94]. The clinical utility of their mechanism of action. The molecular mechanisms, as
α2-adrenoceptor agonists during opioid dose reduction in previously reported, may revert and a second opioid may
patients with OIH has been recently reported to contribute find a different receptor state, thus diminishing the state of
to the recovery of normal nociceptive and anti-nociceptive hyperexcitation [102].
response [95]. Additional studies are necessary to fully char- The most important problem raised in the opioid
acterize the impact of opioid–adrenoceptor agonist coad- switching literature is the conversion ratio among opi-
ministration on analgesia versus undesired adverse effects oids in patients presenting a poor opioid response, with
in the clinical setting. an unfavourable balance between analgesia and adverse
According to the developing understanding of neuroplas- effects. Several studies in the last decade have suggested
ticity observed with the development of tolerance and OIH, that opioids may exert different receptor activities. The
the use of agents that block the activity of NMDA receptors differences in equianalgesic potencies from naïve to highly
may provide an option for the treatment of patients receiv- tolerant patients can be quite profound [107]. Moreover,
ing high doses of opioids who lose their analgesia. NMDA there is no evidence that a specific drug will have more
antagonists may have direct analgesic effects or reverse chance of success when switching from one opioid to
opioid tolerance. Ketamine is a non-competitive NMDA another [107]. It is commonly suggested that the doses
receptor antagonist that exerts its primary effect when the of the different opioids be converted using oral morphine
NMDA receptor-controlled ion channel has been opened by equivalents tables and then reducing the doses by 50% to
a nociceptive barrage. Therefore, ketamine produces only meet the possible asymmetric tolerance between opioids
a weak analgesic effect on acute pain but may significantly [108, 109]. With methadone, the doses should be changed
influence the central hyperexcitability and ‘wind-up’ phe- according to the doses of the previous opioids: the higher
nomena in spinal cord neurons that presumably participate in the dose, the higher the conversion ratio, and the lower the
the development of opioid tolerance and OIH [96]. A burst dose of methadone. This approach has been suggested after
952 S. Mercadante et al.

a series of studies that demonstrated that there is a pro- 2. Johannes CB, Le TK, Zhou X, Johnston JA, Dworkin RH. The
portional inverse ratio with the doses of hydromorphone prevalence of chronic pain in United States adults: results of an
Internet-based survey. J Pain. 2010;11:1230–9.
and morphine, given before switching to methadone. 3. Mercadante S, Portenoy RK. Opioid poorly-responsive cancer
This statement is explained by the peculiar pharmacody- pain: part 2: basic mechanisms that could shift dose response for
namic and pharmacokinetic characteristics of methadone, analgesia. J Pain Symptom Manag. 2001;21:255–64.
including a weak anti-NMDA activity. Continuous use of 4. Cahill CM, Walwyn W, Taylor AMW, Pradhan AAA, Evans CJ.
Allostatic mechanisms of opioid tolerance beyond desensitization
methadone, titrating by one-third of the calculated dose for and downregulation. Trends Pharmacol Sci. 2016;37:963–76.
3 days has been suggested [110]. An alternative proposal 5. Gulur P, Williams L, Chaudhary S, Koury K, Jaff M. Opioid
is an initial fixed ratio of 1:5 with oral morphine equiva- tolerance—a predictor of increased length of stay and higher
lents [111, 112]. Methadone doses should be changed in readmission rates. Pain Physician. 2014;17:E503–7.
6. Volkow ND, McLellan AT. Opioid abuse in chronic pain—
the subsequent days according to the clinical response. misconceptions and mitigation strategies. N Engl J Med.
This approach is based on the individual response, due to 2016;374:1253–63.
a huge number of unpredictable factors, requiring clini- 7. Siegel S, Hinson RE, Krank MD, McCully J. Heroin ‘overdose’
cal flexibility, and provides rapid achievement of clinical death: contribution of drug-associated environmental cues. Sci-
ence. 1982;216:436–7.
stabilization. However, in patients receiving high doses 8. Voon P, Hayashi K, Milloy MJ, Nguyen P, Wood E, Montaner J,
of opioids, switching requires an appropriate setting and et al. Pain among high-risk patients on methadone maintenance
monitoring. In particular, in patients suspected to have treatment. J Pain. 2015;16:887–94.
developed OIH, the conversion ratio should be more cau- 9. Gupta S. Hyperalgesia induced by opioid drugs. J Ration Phar-
macother Res. 2018;4:22–30.
tious. Alternately, in such circumstances, methadone can 10. Silverman SM. Opioid induced hyperalgesia: clinical implica-
be given as needed. tions for the pain practitioner. Physician. 2009;12:679–84.
Despite the favourable effects reported with opioid 11. Compton P, Canamar CP, Hillhouse M, Ling W. Hyperalgesia in
switching, the data are based on open studies, most of heroin dependent patients and the effects of opioid substitution
therapy. J Pain. 2012;13:401–9.
which are retrospective or small case series. Unfortunately, 12. Rieb LM, Norman WV, Martin RE, Berkowitz J, Wood E,
in complex pain conditions it is quite difficult to conduct ran- McNeil R, et al. Withdrawal-associated injury site pain (WISP):
domized controlled studies, and a formal approach according a descriptive case series of an opioid cessation phenomenon.
to evidence-based medicine is unlikely. Pain. 2016;157:2865–74.
13. Arout C, Edens E, Ismene L, Petrakis IL, Sofuoglu M. Targeting
opioid-induced hyperalgesia in clinical treatment: neurobiologi-
cal considerations. CNS Drugs. 2015;29:465–86.
14. Wilson-Poe AR, Lau BK, Vaughan CW. Repeated morphine
4 Conclusion treatment alters cannabinoid modulation of GABAergic synaptic
transmission within the rat periaqueductal grey. Br J Pharmacol.
2015;172:681–90.
Any treatment aimed to reduce opioid consumption may be 15. Wilson-Poe AR, Jeong HI, Vaughan CW. Chronic morphine
indicated in where opioid tolerance of OIH exist. Interven- reduces the readily releasable pool of GABA, a presynaptic
tional techniques able to decrease the pain input may allow mechanism of opioid tolerance. J Physiol. 2017;595:6541–55.
16. Lau BK, Vaughan CW. Descending modulation of pain: the
a decrease in opioid dose, thus reverting the mechanisms GABA disinhibition hypothesis of analgesia. Curr Opin Neuro-
producing tolerance or OIH. Intrathecal therapy with local biol. 2014;29:159–64.
anesthetics and a sympathetic block are the most common 17. Chieng B, Christie MJ. Inhibition by opioids acting on mu-recep-
techniques utilized in these circumstances [113, 114]. tors of GABAergic and glutamatergic postsynaptic potentials in
single rat periaqueductal gray neurones in vitro. Br J Pharmacol.
1994;113:303–9.
Compliance with Ethical Standards 18. Vaughan CW, Christie MJ. Presynaptic inhibitory action of
opioids on synaptic transmission in the rat periaqueductal grey
Funding No funding was received for this article. in vitro. J Physiol. 1997;498:463–72.
19. Connor M, Bagley EE, Chieng BC, Christie MJ. β-Arrestin-2
knockout prevents development of cellular mu-opioid recep-
Conflicts of interest Sebastiano Mercadante, Edoardo Arcuri, and tor tolerance but does not affect opioid-withdrawal-related
Angela Santoni have no conflicts of interest to declare related to this adaptations in single PAG neurons. Brit J Pharmacol.
article. 2015;172:492–500.
20. Vanderah TW, Suenaga NM, Ossipov MH, Malan TP, Lai J,
Porreca F. Tonic descending facilitation from the rostral ven-
References tromedial medulla mediates opioid-induced abnormal pain and
antinociceptive tolerance. J Neurosci. 2001;21:279–86.
21. Meng ID, Harasawa I. Chronic morphine exposure increases the
1. Breivik H, Eisenberg E, O’Brien T. The individual and societal proportion of on-cells in the rostral ventromedial medulla in rats.
burden of chronic pain in Europe: the case for strategic prior- Life Sci. 2007;80:1915–20.
itisation and action to improve knowledge and availability of
appropriate care. BMC Public Health. 2013;13:1229.
Opioid-Induced Tolerance and Hyperalgesia 953

22. Ippolito DL, Temkin PA, Rogalski SL, Chavkin C. N-terminal 41. Raghavendra V, Tanga FY, DeLeo JA. Complete Freunds adju-
tyrosine residues within the potassium channel Kir3 modulate vant induced peripheral inflammation evokes glial activation and
GTPase activity of Galphai. J Biol Chem. 2002;277:32692–6. proinflammatory cytokine expression in the CNS. Eur J Neuro-
23. Torrecilla M, Marker CL, Cintora SC, Stoffel M, Williams JT, sci. 2004;20:467–73.
Wickman K. G-protein-gated potassium channels containing 42. Wang X, Loram LC, Ramos K, et al. Morphine activates neuro-
Kir3.2 and Kir3.3 subunits mediate the acute inhibitory effects of inflammation in a manner parallel to endotoxin. Proc Natl Acad
opioids on locus ceruleus neurons. J Neurosci. 2002;22:4328–34. Sci USA. 2012;109:6325–30.
24. Zhao YL, Chen SR, Chen H, Pan HL. Chronic opioid potenti- 43. Little JW, Cuzzocrea S, Bryant L, Esposito E, Doyle T, Rausaria
ates presynaptic but impairs postsynaptic N-methyl-d-aspartic S, et al. Spinal mitochondrial-derived peroxynitrite enhances
acid receptor activity in spinal cords: implications for opioid neuroimmune activation during morphine hyperalgesia and
hyperalgesia and tolerance. J Biol Chem. 2012;287:25073–85. antinociceptive tolerance. Pain. 2013;154:978–86.
25. Price DD, Mayer DJ, Mao J, Caruso FS. NMDA-receptor antago- 44. Salvemini D, Doyle T, Kress M, Nicol G. Therapeutic targeting
nists and opioid receptor interactions as related to analgesia and of the ceramide-to-sphingosine 1-phosphate pathway in pain.
tolerance. J Pain Symptom Manag. 2000;19(1 Suppl.):S7–11. Trends Pharmacol Sci. 2013;34:110–8.
26. Gintzler AR, Chakrabarti S. Chronic morphine-induced plas- 45. You MH, Kim BM, Chen CH, Begley MJ, Cantley LC, Lee TH.
ticity among signalling molecules. Novartis Found Symptom. Death-associated protein kinase 1 phosphorylates NDRG2 and
2004;261:191–3. induces neuronal cell death. Cell Death Differ. 2017;24:238–50.
27. Hull LC, Llorente J, Gabra BH, et al. The effect of protein kinase 46. Muscoli C, Doyle T, Dagostino C, Bryant L, Chen Z, Watkins
C and G protein-coupled receptor kinase inhibition on tolerance LR, et al. Counterregulation of opioid analgesia by glial-derived
induced by mu-opioid agonists of different efficacy. J Pharmacol bioactive sphingolipids. J Neurosci. 2010;30:15400–8.
Exp Ther. 2010;332:1127–35. 47. Grace PM, Gaudet AD, Staikopoulos V, Maier SF, Hutchinson
28. Melief EJ, Miyatake M, Bruchas MR, Chavkin C. Ligand- MR, Salvemini D, et al. Nitroxidative signaling mechanisms in
directed c-Jun N-terminal kinase activation disrupts opioid recep- pathological pain. Trends Neurosci. 2016;39:862–79.
tor signaling. Proc Natl Acad Sci USA. 2010;107:11608–13. 48. Watkins LR, Hutchinson MR, Rice KC, Maier SF. The “toll”
29. Bobeck EN, Ingram SL, Hermes SM, Aicher SA, Morgan MM. of opioid-induced glial activation: improving the clinical effi-
Ligand-biased activation of extracellular signal-regulated kinase cacy of opioids by targeting glia. Trends Pharmacol. Sci.
1/2 leads to differences in opioid induced antinociception and 2009;30:581–91.
tolerance. Behav Brain Res. 2016;298:17–24. 49. Nakamoto K, Kawasaki S, Kobori T, et al. Involvement of matrix
30. Morgan MM, Bobeck EN, Ingram SL. Glutamate modulation metalloproteinase-9 in the development of morphine tolerance.
of antinociception, but not tolerance, produced by morphine Eur J Pharmacol. 2012;683:86–92.
microinjection into the periaqueductal gray of the rat. Brain Res. 50. Hutchinson MR, Zhang Y, Shridhar M, et al. Evidence that opi-
2009;1295:59–66. oids may have toll-like receptor 4 and MD-2 effects. Brain Behav.
31. Xie JY, Herman DS, Stiller C-O, Gardell LR, Ossipov MH, Lai J, Immun. 2010;24:83–95.
et al. Cholecystokinin in the rostral ventromedial medulla medi- 51. Watkins LR, Hutchinson MR, Johnston IN, Maier SF. Glia:
ates opioid- induced hyperalgesia and antinociceptive tolerance. novel counter-regulators of opioid analgesia. Trends Neurosci.
J Neurosci. 2005;25:409–16. 2005;28:661–9.
32. Thomas J, Mustafa S, Johnson J, Nicotra L, Hutchinson M. The 52. Eidson LN, Inoue K, Young LJ, Tansey MG, Murphy AZ. Toll-
relationship between opioids and immune signaling in the spinal like receptor 4 mediates morphine-induced neuroinflammation
cord. Handb Exp Pharmacol. 2015;227:207–38. and tolerance via soluble tumor necrosis factor signaling. Neu-
33. Xu J-TT, Yaster M, Tao Y-XX, et al. Opioid receptor-triggered ropsychopharmacology. 2017;42:661–70.
spinal mTORC1 activation contributes to morphine tolerance and 53. Hutchinson MR, Lewis SS, Coats BD, Skyba DA, Crysdale NY,
hyperalgesia. J Clin Investig 2014;124:592–603. Berkelhammer DL, et al. Reduction of opioid withdrawal and
34. Harada S, Nakamoto K, Tokuyama S. The involvement of mid- potentiation of acute opioid analgesia by systemic AV411 (ibudi-
brain astrocyte in the development of morphine tolerance. Life last). Brain Behav Immun. 2009;23:240–50.
Sci. 2013;93:573–8. 54. Johnston IN, Milligan ED, Wieseler-Frank J, et al. A role for pro-
35. Eidson LN, Murphy AZ. Blockade of toll-like receptor 4 attenu- inflammatory cytokines and fractalkine in analgesia, tolerance,
ates morphine tolerance and facilitates the pain relieving proper- and subsequent pain facilitation induced by chronic intrathecal
ties of morphine. J Neurosci. 2013;33:15952–63. morphine. J Neurosci. 2004;24:7353–65.
36. Beutler B, Du X, Poltorak A. Identification of Toll-like receptor 55. Johnson JL, Rolan PE, Johnson ME, Bobrovskaya L, Williams
4 (Tlr4) as the sole conduit for LPS signal transduction: genetic DB, Johnson K, et al. Codeine-induced hyperalgesia and allo-
and evolutionary studies. J Endotoxin Res. 2001;7:277–80. dynia: investigating the role of glial activation. Transl Psychiatry.
37. Thomas J, Mustafa S, Johnson J, Nicotra L, Hutchinson M. The 2014;4:e482.
relationship between opioids and immune signaling in the spinal 56. Gruber-Schoffnegger D, Drdla-Schutting R, Honigsperger C,
cord. Handb Exp Pharmacol. 2015;227:207–38. Wunderbaldinger G, Gassner M, Sandkuhler J. Induction of
38. Tanga FY, Raghavendra V, DeLeo JA. Quantitative real-time thermal hyperalgesia and synaptic long-term potentiation in the
RT-PCR assessment of spinal microglial and astrocytic activa- spinal cord lamina I by TNF-alpha and IL-1beta is mediated
tion markers in a rat model of neuropathic pain. Neurochem Int. by glial cells. J Neurosci. 2013;33:6540–51.
2004;45:397–407. 57. Melik Parsadaniantz S, Rivat C, Rostene W, Reaux-Le Goazigo
39. Tanga FY, Nutile-McMenemy N, DeLeo JA. The CNS role of A. Opioid and chemokine receptor crosstalk: a promising target
Toll-like receptor 4 in innate neuroimmunity and painful neu- for pain therapy? Nat Rev Neurosci. 2015;16:69–78.
ropathy. Proc Natl Acad Sci USA. 2005;102:5856–61. 58. Zhang N, Rogers TJ, Caterina M, Oppenheim JJ. Proinflamma-
40. Lewis SS, Hutchinson MR, Rezvani N, Loram LC, Zhang Y, tory chemokines, such as C-C chemokine ligand 3, desensitize
Maier SF, et al. Evidence that intrathecal morphine-3-glucuro- mu-opioid receptors on dorsal root ganglia neurons. J Immu-
nide may cause pain enhancement via toll-like receptor 4/MD-2 nol. 2004;173:594–9.
and interleukin-1beta. Neuroscience. 2010;165:569–83.
954 S. Mercadante et al.

59. Rivat C, Sebaihi S, Van Steenwinckel J, et al. Src family hyperalgesia in mice lacking the nociception precursor polypep-
kinases involved in CXCL12-induced loss of acute morphine tide or the nociceptin receptor. Eur J Neurosci. 2003;17:2381–7.
analgesia. Brain Behav Immun. 2014;38:38–52. 79. Rizzi A, Nazzaro C, Marzola GG, et al. Endogenous nociceptin/
60. Lin CP, Kang KH, Lin TH, et al. Role of spinal CXCL1 orphanin FQ signalling produces opposite spinal antinociceptive
(GROalpha) in opioid tolerance: a human-to-rodent transla- and supraspinal pronociceptive effects in the mouse formalin test:
tional study. Anesthesiology. 2015;122:666–76. pharmacological and genetic evidences. Pain. 2006;124:100–8.
61. Luo X, Wang X, Xia Z, Chung SK, Cheung CW. CXCL12/ 80. Garcia-Recio S, Gascón P. Biological and pharmacological
CXCR61 axis: an emerging neuromodulator in pathological aspects of the NK1-receptor. Biomed Res Int. 2015;2015:495704.
pain. Rev Neurosci. 2016;27:83–92. 81. Tumati S, Largent-Milnes TM, Keresztes AI, et al. Tachykinin
62. Takayama N, Ueda H. Morphine-induced chemotaxis and NK(1) receptor antagonist co-administration attenuates opioid
brain-derived neurotrophic factor expression in microglia. J withdrawal-mediated spinal microglia and astrocyte activation.
Neurosci. 2005;25:430–5. Eur J Pharmacol. 2012;684:64–70.
63. Horvath RJ, Romero-Sandoval EA, De Leo JA. Inhibition of 82. Liang DY, Li X, Clark JD. Epigenetic regulation of opioid-
microglial P2X4 receptors attenuates morphine tolerance, Iba1, induced hyperalgesia, dependence, and tolerance in mice. J Pain.
GFAP and mu opioid receptor protein expression while enhanc- 2013;14:36–47.
ing perivascular microglial ED2. Pain. 2010;150:401–13. 83. Doehring A, Geisslinger G, Lotsch J. Epigenetics in pain and
64. Zhou D, Chen ML, Zhang YQ, Zhao ZQ. Involvement of spinal analgesia: an imminent research field. Eur J Pain. 2011;15:11–6.
microglial P2X7 receptor in generation of tolerance to mor- 84. Doehring A, Oertel BG, Sittl R, Lotsch J. Chronic opioid use
phine analgesia in rats. J Neurosci. 2010;30:8042–7. is associated with increased DNA methylation correlating with
65. Chen ML, Cao H, Chu YX, et al. Role of P2X7 receptor- increased clinical pain. Pain. 2013;154:15–23.
mediated IL-18/IL-18R signaling in morphine tolerance: Mul- 85. Viet CT, Dang D, Aouizerat BE, Miaskowski C, Ye Y, Viet DT,
tiple glial-neuronal dialogues in the rat spinal cord. J Pain. et al. OPRM1 methylation contributes to opioid tolerance in can-
2012;13:945–58. cer patients. J Pain. 2017;18:1046–59.
66. Ferrini F, Trang T, Mattioli TA. Morphine hyperalgesia gated 86. Chao YC, Xie F, Li X, Guo R, Yang N, Zhang C, et al. Demethyl-
through microglia-mediated disruption of neuronal ­Cl– homeo- ation regulation of BDNF gene expression in dorsal root ganglion
stasis. Nat Neurosci. 2013;16:183–92. neurons is implicated in opioid-induced pain hypersensitivity in
67. Mattioli TA, Leduc-Pessah H, Skelhorne-Gross G, et al. Toll- rats. Neurochem Int. 2016;97:91–8.
like receptor 4 mutant and null mice retain morphine-induced 87. He Y, Wang ZJ. Let-7 microRNAs and opioid tolerance. Front
tolerance, hyperalgesia, and physical dependence. PLoS One. Genet. 2012;3:110.
2014;9:e97361. 88. He Y, Yang C, Kirkmire CM, Wang ZJ. Regulation of opioid
68. Grace PM, Hutchinson MR, Maier SF, Watkins LR. Patho- tolerance by let-7 family microRNA targeting the mu opioid
logical pain and the neuroimmune interface. Nat Rev Immunol. receptor. J Neurosci. 2010;30:10251–8.
2014;14:217–31. 89. Wu Q, Zhang L, Law PY, Wei LN, Loh HH. Long-term mor-
69. Ossipov MH, Lai J, Vanderah TW, Porreca F. Induction of pain phine treatment decreases the association of mu-opioid recep-
facilitation by sustained opioid exposure: relationship to opioid tor (MOR1) mRNA with polysomes through miRNA23b. Mol
antinociceptive tolerance. Life Sci. 2003;73:783–800. Pharmacol. 2009;75:744–50.
70. Toll L, Bruchas MR, Calo G, Cox BM, Zaveri NT. Nocic- 90. Mercadante S. Portenoy RK Opioid poorly-responsive cancer
eptin/orphanin FQ receptor structure, signaling, ligands, func- pain. Part 3. Clinical strategies to improve opioid responsiveness.
tions, and interactions with opioid systems. Pharmacol Rev. J Pain Symptom Manag. 2001;21:338–54.
2016;68:419–57. 91. Edwards HL, Mulvey MR, Bennett MI. Cancer-related neuro-
71. Faris PL, Komisaruk BR, Watkins LR, Mayer DJ. Evidence for pathic pain. Cancers (Basel). 2019;11(3):373.
the neuropeptide cholecystokinin as an antagonist of opiate anal- 92. Lee JT, Sanderson CR, Xuan W, Agar M. Lidocaine for cancer
gesia. Science. 1983;219:310–2. pain in adults: a systematic review and meta-analysis. J Palliat
72. Rothman RB. A review of the role of anti-opioid peptides in Med. 2019;22:326–34.
morphine tolerance and dependence. Synapse. 1992;12:129–38. 93. Mercadante S, Fulfaro F, Casuccio A. A randomized controlled
73. McNally GP. Pain facilitatory circuits in the mammalian cen- study on the use of anti-inflammatory drugs in patients with can-
tral nervous system: their behavioral significance and role cer pain on morphine therapy: effects on dose escalation and a
in morphine analgesic tolerance. Neurosci Biobehav Rev. pharmacoeconomic analysis. Eur J Cancer. 2002;38:1358–66.
1999;23:1059–78. 94. Chabot-Doré AJ, Schuster DJ, Stone LS, Wilcox GL. Analgesic
74. Dourish CT, O’Neill MF, Schaffer LW, Siegl PK, Iversen SD. synergy between opioid and α2-adrenoceptors. Br J Pharmacol.
The cholecystokinin receptor antagonist devazepide enhances 2015;172:388–402.
morphine-induced analgesia but not morphine-induced respira- 95. Belgrade M, Hall S. Dexmedetomidine infusion for the
tory depression in the squirrel monkey. J Pharmacol Exp Ther. management of opioid-induced hyperalgesia. Pain Med.
1990;255:1158–65. 2010;11:1819–26.
75. Hoffmann O, Wiesenfeld-Hallin Z. The CCK-B receptor antago- 96. Mercadante S. The patient with difficult cancer pain. Cancers
nist Cl 988 reverses tolerance to morphine in rats. NeuroReport. (Basel). 2019;11(4):565.
1994;5:2565–8. 97. Mercadante S, Caruselli A, Casuccio A. The use of ketamine in
76. Simonin F, Schmitt M, Laulin J-P, et al. RF9, a potent and a palliative-supportive care unit: a retrospective analysis. Ann
selective neuropeptide FF receptor antagonist, prevents opioid- Palliat Med. 2018;7:205–10.
induced tolerance associated with hyperalgesia. Proc Natl Acad 98. Pasternak GW. Molecular biology of opioid analgesia. J Pain
Sci USA. 2006;103:466–71. Symptom Manag. 2005;19:S2.
77. Elhabazi K, Trigo JM, Mollereau C, et al. Involvement of neu- 99. Connor M, Osborne PB, Christies MJ. Mu-opioid recep-
ropeptide FF receptors in neuroadaptive responses to acute and tor desensitization: is morphine different? Br J Pharmacol.
chronic opiate treatments. Br J Pharmacol. 2012;165:424–35. 2004;143:685–96.
78. Depner UB, Reinscheid RK, Takeshima H, Brune K, Zeilhofer
HU. Normal sensitivity to acute pain, but increased inflammatory
Opioid-Induced Tolerance and Hyperalgesia 955

100. Hashimoto T, Saito Y, Yamada K, et al. Enhancement of mor- 109. Dale O, Moksnes K, Kaasa S. European Palliative Care Research
phine analgesic effect with induction of mu-opioid receptor endo- Collaborative pain guidelines: opioid switching to improve anal-
cytosis in rats. Anesthesiology. 2006;105:574–80. gesia or reduce side effects. A systematic review. Palliat Med.
101. Kieffer BL, Evans CJ. Opioid tolerance—in search of the holy 2011;25:494–503.
grail. Cell. 2002;108:587–90. 110. Bruera E, Pereira J, Watanabe S, Belzile M, Kuehn N, Hanson
102. Mercadante S, Bruera E. Opioid switching in cancer pain: J. Opioid rotation in patients with cancer pain. A retrospective
from the beginning to nowadays. Crit Rev Oncol Hematol. comparison of dose ratios between methadone, hydromorphone,
2016;99:241–8. and morphine. Cancer. 1996;78:852–7.
103. Mercadante S. Managing difficult pain conditions in the cancer 111. Mercadante S, Casuccio A, Calderone L. Rapid switching from
patient. Curr Pain Headache Rep. 2014;18:395. morphine to methadone in cancer patients with poor response to
104. Mercadante S. Switching methadone: a 10-year experience morphine. J Clin Oncol. 1999;17:3307–12.
of 345 patients in an acute palliative care unit. Pain Med. 112. Mercadante S, Bianchi M, Villari P, Ferrera P, Casuccio A, Ful-
2012;13:399–404. faro F, et al. Opioid plasma concentration during switching from
105. Reddy A, Yennurajalingam S, Pulivarthi K, Palla SL, Wang X, morphine to methadone: preliminary data. Support Care Cancer.
Kwon JH, et al. Frequency, outcome, and predictors of success 2003;11:326–31.
within 6 weeks of an opioid rotation among outpatients with 113. Kurita GP, Sjøgren P, Klepstad P, Mercadante S. Interventional
cancer receiving strong opioids. Oncologist. 2013;18:212–20. techniques to management of cancer-related pain: clinical and
106. Mercadante S, Arcuri E. Hyperalgesia and opioid switching. Am critical aspects. Cancers (Basel). 2019;11(4):443.
J Hosp Palliat Care. 2005;22:291–4. 114. Mercadante S, Klepstad P, Kurita GP, Sjøgren P, Giarratano
107. Prommer E. Rotating methadone to other opioids: a lesson in A. Sympathetic blocks for visceral cancer pain management: a
the mechanisms of opioid tolerance and opioid-induced pain. J systematic review and EAPC recommendations. Crit Rev Oncol
Palliat Med. 2006;9:488–93. Hematol. 2015;96:577–83.
108. Mercadante S, Caraceni A. Conversion ratios for opioid switch-
ing in the treatment of cancer pain: a systematic review. Palliat
Med. 2011;25:494–503.

You might also like