Acute and Neurophatic Pain

download Acute and Neurophatic Pain

of 23

Transcript of Acute and Neurophatic Pain

  • 7/30/2019 Acute and Neurophatic Pain

    1/23

    6.14 Acute and Neuropathic PainP Honore and M F Jarvis, Abbott Laboratories, Abbott Park, IL, USA

    & 2007 Elsevier Ltd. All Rights Reserved.

    6.14.1 Introduction: Pain States 328

    6.14.2 Neuropathophysiology of Pain 328

    6.14.3 Experimental Pain Models 331

    6.14.3.1 Models of Acute Pain 331

    6.14.3.1.1 Acute thermal pain 331

    6.14.3.1.2 Acute mechanical pain 332

    6.14.3.1.3 Acute chemical pain 332

    6.14.3.2 Models of Nociceptive Pain 333

    6.14.3.2.1 Adjuvant-induced arthritis 333

    6.14.3.2.2 Unilateral inflammation 333

    6.14.3.2.3 Models of osteoarthritic pain 333

    6.14.3.3 Models of Neuropathic Pain 334

    6.14.3.3.1 Direct trauma to nerves 3346.14.3.3.2 Inflammation/neuritis/nerve compression 334

    6.14.3.3.3 Diabetes 335

    6.14.3.3.4 Chemotherapy-induced neuropathic pain (vincristine/paclitaxel/platine) 335

    6.14.4 Clinical Trial Issues 335

    6.14.4.1 Translational Medicine in Pain 337

    6.14.5 Current Treatments 337

    6.14.5.1 Opioids 338

    6.14.5.2 Anti-Inflammatory Drugs 338

    6.14.5.2.1 Nonsteroidal anti-inflammatory drugs 338

    6.14.5.2.2 Cyclooxygenase-2 inhibitors 339

    6.14.5.3 Analgesic Adjuvants 3396.14.5.3.1 Amitriptyline 339

    6.14.5.3.2 Antiepileptics 339

    6.14.5.3.3 Pregabalin 339

    6.14.5.3.4 Antidepressants 339

    6.14.6 Unmet Medical Needs 340

    6.14.7 New Research Areas 340

    6.14.7.1 Neuronal Nicotinic Receptor Agonists 340

    6.14.7.2 Vanilloid Receptor Modulators 340

    6.14.7.3 Excitatory Amino Acid Receptor Antagonists 341

    6.14.7.4 Calcium Channel Modulators 342

    6.14.7.5 Cannabinoids 342

    6.14.7.6 Sodium Channel Modulators 343

    6.14.7.7 Purines 344

    6.14.7.7.1 P1 receptor agonists 344

    6.14.7.7.2 P2 receptor antagonists 344

    6.14.7.8 Emerging Pain Targets 345

    6.14.8 Conclusions 346

    References 346

    327

  • 7/30/2019 Acute and Neurophatic Pain

    2/23

    6.14.1 Introduction: Pain States

    The International Association for the Study of Pain (IASP) defines pain as an unpleasant sensory and emotional

    experience associated with actual or potential tissue damage, or described in terms of such damage.1 This definition

    clearly indicates that pain is a multidimensional experience. Pain is not homogenous and can be classified temporally as

    acute or chronic. Acute or physiological pain is an early warning against potential injury, a vital defense mechanism,

    whereas chronic pain does not play any useful role. As such, chronic pain can be very detrimental to the quality of life of

    an individual, disrupting sleep and normal living, and degrading health and functional capability.2

    Chronic pain is one of the most common complaints for which individuals solicit medical attention. It can affect

    general and psychological health (as is evident by the high degree of comorbidity of chronic pain and emotional

    disorders such as depression and anxiety), and can also have deleterious socioeconomic consequences since it is often

    associated with the loss of work time and an increased use of healthcare resources. Published estimates of the

    prevalence of chronic pain typically range from 2% to 45% with 50% of respondents reportedly suffering from chronic

    pain.3 Interestingly, there was no significant difference between genders and, as might be expected, the proportion and

    the degree of pain significantly increased with age. Furthermore, 3340% of chronic pain patients included in their

    study were unhappy with medical examinations, medical tests, and treatments related to their chronic pain state. 4 This

    lack of satisfactory pain relief in chronic pain patients was also identified in a 2004 Americans Living with Pain Survey

    conducted on behalf of the American Chronic Pain Association.5 In this study, 50% of the chronic pain patients

    surveyed felt that their pain was not under control. In the face of this growing unmet medical need is an increasing

    awareness of undertreated pain resulting in a more aggressive use of analgesics, especially opioids. As the population ofelderly people continues to increase the demand for therapies to treat arthritis, pain associated with osteoporosis, and

    other painful diseases of the aged will also increase. These results clearly illustrate the need for better and more

    efficacious pain management medications, programs, and therapies. One of the greatest challenges in creating more

    efficacious medications for pain control has been the heterogeneity of the condition itself, including: the causes and

    underlying pathologies; the redundancies in pain perception; and the usefulness of current pharmacological therapies.

    6.14.2 Neuropathophysiology of Pain

    The conceptualization of the neurobiology of pain has undergone continuous refinement with increasing knowledge of

    multiple nociceptive targets and pathways.6,7 The psychophysical parameters used to describe nociceptive processing

    have thus been refined to differentiate acute withdrawal behaviors in response to dangerous (e.g., sharp or hot stimuli)stimuli in the environment (acute nociception) from increased sensitivity to mildly painful stimuli (hyperalgesia) or

    to otherwise innocuous stimuli (allodynia) (Figure 1).8 An increase in stimulus intensity in any sensory modality will

    eventually become noxious (Figure 1). Obviously, the sensation of noxious environmental stimuli (acute pain) is

    physiologically protective. However, following injury, this psychophysical function shifts such that previous noxious

    stimuli are now perceived as exceedingly painful (hyperalgesia). Additionally, tissue injury results in ongoing

    or spontaneous pain and the perception that normally nonnoxious stimuli are pain generating (allodynia). It is now

    Hyperalgesia

    Allodynia

    Innocuous Noxious

    Stimulus intensity

    Pain

    sensation

    0

    100

    50 Injury Normal

    Low High

    Figure 1 Psychophysical representation of hyperalgesia and allodynia sensory sensitivity. Following injury, being tissue or

    nerve injury, pain transmission and perception are changed so that a normally painful stimulus is going to be felt more painful

    (hyperalgesia) and a normally nonpainful stimulus is going to be felt as painful (allodynia). (Adapted from Cervero, F.; Laird, J. M.,

    Pain 1996, 68, 1323.)

    328 Acute and Neuropathic Pain

  • 7/30/2019 Acute and Neurophatic Pain

    3/23

    well appreciated that distinct sensory mechanisms contribute to physiological pain, to pain arising from tissue

    damage (inflammatory or nociceptive pain), and to pain arising from injury to the nervous system (neuropathic

    pain).9

    Nociceptive pain is caused by the ongoing activation of A-d and C-nociceptors in response to a noxious stimulus

    (injury, disease, inflammation) (Figure 2). It can be further classified into visceral pain (deep cramping sensation

    associated with referred pain), superficial somatic pain (skin; well-localized sharp, pricking, or burning sensation), and

    deep somatic pain (muscle, joint capsules, and bone; diffuse dull or aching sensation). Under normal physiological

    conditions, there is a close correspondence between pain perception and stimulus intensity, and the sensation of pain is

    indicative of real or potential tissue damage. As the nervous system becomes sensitized (responding more strongly than

    normal to peripheral stimuli), in addition to spontaneous pain, nociceptive pain is also associated with evoked

    hyperalgesic and allodynic conditions.1012 In general, nociceptive pain abates completely upon the resolution of injury

    if the disease process is controlled. Because of this, the use of disease-modifying therapies is being emphasized in the

    treatment of nociceptive chronic pain as illustrated in the treatment of rheumatoid arthritis not only by

    antiinflammatory agents but also by biological therapies such as tumor necrosis factor-a (TNF-a) antagonists.13

    Unlike nociceptive pain, neuropathic pain can persist long after the initiating injurious event has been removed and

    any damage has healed. This then leads to abnormal processing of sensory information by the nervous system.

    Neuropathic pain can be classified as peripheral (painful peripheral mononeuropathy and polyneuropathy) or central

    (post stroke, following spinal cord injury) and can originate from nerve injury following a wide array of conditions/

    events, e.g., direct trauma to nerves, inflammation/neuritis/nerve compression, diabetes, infections (herpes zoster,

    human immunodeficiency virus (HIV)), tumors (nerve compression/infiltration), toxins (chemotherapy), and primary

    neurological diseases.14,15 Following nerve injury, changes occur in the central nervous system (CNS) that can persist

    indefinitely. Under these conditions of sensitization, pain can occur without a specific stimulus or can be

    disproportionate to the stimulus intensity. The sensation of neuropathic pain may also be constant or intermittent and

    is felt in many different ways (e.g., allodynia or hyperalgesia associated with mechanical or thermal stimuli but also

    spontaneous sensations such as burning, tingling, prickling, shooting, deep aching, and spasm).14,15

    Descendinginhibitions

    Nociceptors

    A fibers A fibers

    Brain

    Ascendingpain

    pathways

    Pathophysiological pain

    Peripheralsensitization

    Nociceptors

    Descendinginhibitions

    Ascendingpain

    pathways

    Brain

    Centralsensitization

    Spinal cord

    Sprouting

    Normal pain

    Spinal cord

    Figure 2 Normal and pathophysiological transmission of pain. Under normal conditions, pain stimuli such as noxious heat are

    transmitted from the peripheral site (e.g., skin or joints) through the nociceptive primary afferent fibers to the spinal cord and the

    brain. Pathophysiological conditions are associated with peripheral and central sensitization. Peripheral sensitization can result

    from the sensitization of nociceptors by inflammatory mediators, neurotrophic factors released during tissue damage or by

    inflammatory cells. Peripheral sensitization is also associated with intense, repeated, or prolonged action potential generation in

    primary sensory afferents that is mediated by altered expression and activity of voltage-gated sodium and calcium channels.

    Consequences of peripheral sensitization are a lowering of the activation threshold of nociceptors and an increase in their firing

    rate. These changes result in the production of hyperalgesia and allodynia associated with nociceptive chronic pain. Central

    sensitization (long-lasting increases in dorsal horn neuron excitability and responsiveness) is associated with spontaneous

    dorsal horn neuron activity, responses from neurons that normally only respond to low intensity stimuli (altered neural

    connections following sprouting of Ab fibers to superficial laminae) and reduction in central inhibition. Central sensitization is

    associated with persistent pain, hyperalgesia, allodynia, and the spread of pain to uninjured tissue. In addition, it reflects a

    complex series of changes occurring in the spinal cord that may promote long-lasting increases in dorsal horn neuron

    excitability including the involvement of astrocytes and microglia activation.

    Acute and Neuropathic Pain 329

  • 7/30/2019 Acute and Neurophatic Pain

    4/23

    Tissue injury results in the release of pronociceptive mediators that sensitize peripheral nerve terminals (peripheral

    sensitization), leading to phenotypic alterations of sensory neurons and increased excitability of spinal cord dorsal horn

    neurons (central sensitization).7,9 Nerve injury may be associated with abnormal firing of the injured neurons, leading also

    to central sensitization and phenotypic changes in spinal cord neurons.16,17 In addition, descending supraspinal systems

    modulate nociceptive responses.18 A multitude of receptors, transmitters, second messenger systems, transcription

    factors, and other signaling molecules are now appreciated to be involved in pain pathways (Figures 2 and 3).6

    As noted above, two mechanisms play a key role in the development and maintenance of chronic pain, namely

    peripheral and central sensitization.19 Peripheral sensitization can result from the sensitization of nociceptors by

    inflammatory mediators (e.g., prostaglandin E2 (PGE2), serotonin (5HT), bradykinin, epinephrine, adenosine), by

    neurotrophic factors released during tissue damage (e.g., nerve growth factor (NGF)) or by inflammatory cells

    (proinflammatory cytokines including interleukin-1 (IL1)). Peripheral sensitization is also associated with intense,

    repeated, or prolonged action potential generation in primary sensory afferents that is mediated by altered expression

    and activity of voltage-gated sodium and calcium channels.6 Consequences of peripheral sensitization are a lowering of

    the activation threshold of nociceptors and an increase in their firing rate. These changes result in the production of

    hyperalgesia and allodynia associated with nociceptive chronic pain. In addition, peripheral sensitization plays also an

    important role in the development and maintenance of central sensitization.6,17

    Central sensitization (long-lasting increases in dorsal horn neuron excitability and responsiveness) is associated with

    spontaneous dorsal horn neuron activity, responses from neurons that normally only respond to low-intensity stimuli

    (altered neural connections following sprouting of Ab fibers to superficial laminae), expansion of dorsal horn neuron

    receptive fields, and reduction in central inhibition.2022 Central sensitization is associated with persistent pain,

    hyperalgesia, allodynia, and the spread of pain to uninjured tissue, i.e., secondary hyperalgesia due to increased receptor

    Dorsal root ganglionprimary afferent cell body

    A

    SPBDNFGALNPYNav1.3

    C

    SPCGRP

    TRPV1ASICs

    Nav1.8

    Peripheral terminalprimary afferent fiber

    P2X3 Nav1.3

    TRPV1

    mGLU

    TRPs

    opioid

    ASICs

    TNFIL1

    Spinal cord

    Nav1.8

    TRPs

    P2X3

    opioid NK-1

    NMDA

    2

    GABA-BCCKB

    A1

    mGLU

    CGRP-1

    Glia

    Central terminalprimary afferent fiber

    Post

    synaptic

    neuron

    SP

    NE5HT

    GABA

    ENK

    GLU

    ACh

    CGRP

    Descending systemsor

    interneurons

    Figure 3 Pain transmission sites can be simply divided into two: the peripheral compartment (e.g., skin, muscle, organs) that

    encompasses primary afferent fibers and dorsal root ganglions and the central compartment that includes the spinal cord and

    brain. A multitude of receptors, transmitters, second messenger systems, transcription factors, and other signaling molecules

    located all along pain transmission pathways are now appreciated to be involved in pain signaling. Tissue injury results in the

    release of pronociceptive mediators that activate and sensitize peripheral nerve terminals (peripheral sensitization) through

    various receptors/channels, leading to phenotypic alterations of sensory neurons with changes of receptor expression in DRG

    cell bodies and changes in neurotransmitter contents and increased excitability of spinal cord dorsal horn neurons (central

    sensitization) due to a variety of changes in receptor expression patterns and neurotransmitter release. In addition, interneurons

    and descending supraspinal systems modulate nociceptive responses either through excitatory or inhibitory effects. 5HT

    (serotonin), A1 (adenosine 1), a2 (alpha 2 adrenergic), ACH (acetylcholine), ASICs (acid sensing ion channels), BDNF (brain

    derived nerve growth factor), CCKB (cholecystokinin), CGRP (calcitonin gene related peptide), ENK (enkephalines), GABA, GAL

    (galanin), GLU (glutamate), mGlu (metabotropic glutamate receptors), Nav1.3 (sodium channels), Nav1.8 (sodium channels),

    NE (norepinephrine), NK-1 (neurokinin-1 receptor), NMDA (N-methyl-D-aspartate receptor), NPY (neuropeptide Y), opioid, P2X3

    (ATP receptor), SP (substance P), TRP (transient receptor potential), and TRPV1 (vanilloid receptor 1).

    330 Acute and Neuropathic Pain

  • 7/30/2019 Acute and Neurophatic Pain

    5/23

    field of spinal neurons. In addition, it reflects a complex series of changes occurring in the spinal cord that may promote

    long-lasting increases in dorsal horn neuron excitability. This process is also know as wind-up in that the response of

    sensitized dorsal horn neurons is exaggerated relative to the normal situation.2022 While both peripheral and central

    sensitization play a role in nociceptive chronic pain, central sensitization clearly plays a key role in neuropathic pain.

    Thus, central sensitization also explains the observation that established pain is more difficult to suppress than acute

    pain because of the maladaptive changes that have taken place in the CNS.6,17 Interestingly, not only neurons, but also

    glia, e.g. astrocytes and microglia, as well as infiltrating mast cells are involved in the generation and maintenance of

    central sensitization.23,24

    In addition to the activation of pronociceptive inflammatory and/or neurotrophic messengers noted above, the

    sensitization of the nervous system in response to chronic pain involves the alteration and/or activation of many

    neurotransmitter systems that have been extensively reviewed elsewhere.6,15 Chronic pain is mediated by altered

    neuronal excitability involving activation of sodium and calcium channels in both peripheral and spinal neurons.

    Additionally, there is enhanced glutamatergic activity and a concomitant decrease in GABAergic inhibitory

    neuromodulation at the level of the dorsal spinal horn.6,15 This altered neurochemical profile contributes to the

    heightened state of neuronal excitability (e.g., wind-up) and can be viewed as a shift in the balance of excitatory and

    inhibitory systems that also incorporates activation of intracellular signaling cascades (e.g., ras-mitogen-activated protein

    kinase (MAPK) pathway) and recruitment of neurotrophic neuropeptides including substance P, neuropeptide Y,

    and brain-derived neurotrophic factor (BDNF). These changes in spinal neuron neurochemistry are also accompanied

    by upregulation of specific excitatory amino acid receptors (e.g., a-amino-3-hydroxy-5-methylisoxazole-4-propionic acid

    (AMPA) and N-methyl-D-aspartate (NMDA)), as well as increased calcium and potassium ion channel activity. Taken

    together, chronic pain is associated with a large variety of deranged patterns of neurotransmission at multiple levels of

    the neuroaxis with considerable target and pathway redundancy. Thus, in the absence of ongoing injury, chronic pain can

    be viewed as a disease in itself. The enhanced appreciation of the many neurochemical and neurophysiological

    alterations in neuronal function associated with chronic pain has led to the development of both new preclinical models

    of pain and a variety of potentially useful therapeutic interventions.

    6.14.3 Experimental Pain Models

    To facilitate the study of pain transmission and the characterization of novel analgesic compounds, an array of

    experimental animal pain models has been developed mainly in rodents, reflecting all types of pain, from acute to

    chronic, somatic to visceral, and nociceptive to neuropathic and cancer-related pain. Depending on the model, painmeasurements can encompass spontaneous pain behaviors as well as pain evoked by various sensory modalities. It is

    important to note that in rodents, measuring spontaneous pain is very difficult and is generally limited to the

    observation of quantifiable nocifensive (pain-escape) behaviors such as hind paw lifting or altered grooming. However,

    experimental measures of evoked pain are well characterized and are analogous to clinical diagnostic methods. In the

    following overview, acute pain refers to pain that lasts from seconds to a day while chronic pain typically refers to

    experimental pain manipulations that persist for at least several days. In addition, this section will focus on the most-

    widely used preclinical pain models.

    The majority of these animal models of pain were originally developed in rats. Except as noted below, essentially all

    have also been successfully carried out using various mouse strains including gene-disrupted (knockout) mice.25

    However, significant differences in the basal nociceptive sensitivity and analgesic response have been noted for

    different mouse strains serving to further complicate the interpretation of the knockout phenotype. 26

    6.14.3.1 Models of Acute Pain

    Animal models of acute pain allow the evaluation of the effects of potential analgesics on pain sensation/transmission in

    an otherwise normal animal. In addition, the same tests may be used to measure stimulus-evoked pain in animals with

    chronic inflammation or nerve injury. Usually, these tests rely on an escape behavior/withdrawal reflex or vocalization as

    an index of pain. The animals have control over the duration of the pain, that is, their behavioral response leads to

    termination of the noxious stimulus.

    6.14.3.1.1 Acute thermal pain

    Models have been developed to interrogate acute thermal pain sensitivity, using various means of applying a noxious

    heat stimulus to the paw or the tail of rodents. These models have been widely used in the characterization of opioid

    analgesics. Usually, latency to behavioral response is recorded and a cutoff is set to avoid any tissue damage to the

    Acute and Neuropathic Pain 331

  • 7/30/2019 Acute and Neurophatic Pain

    6/23

    animal. The tail-flick test involves the application of a focused heat (usually light) source on the tail until a tail-flick

    (rapid removal of the tail) reflex occurs. This test has an advantage in that it does not involve repeated assessments of

    animal behavior, i.e., animals learning with time when the stimulus is going to be applied and anticipating the test.

    The hot plate assay uses a hot plate set at a fixed temperature, usually 5055 1C. Latency to licking, shaking of hind

    limbs or fore limbs, in addition to latency to jump can be recorded and statistically analyzed for groups of animals. This

    assay can be difficult to standardize since the heat stimulus is not delivered in a controlled fashion. Possible sources of

    variability include differential exposure to the heated plat depending on how much weight the animal puts on each limb.

    Another approach to assess acute thermal pain is the use of a radiant heat source.27 Using this methodology, the

    temperature of the heat source applied to the hind paw increases over time until it reaches a painful threshold. Latency

    to hind paw withdraw is recorded and analyzed. In each test session, each animal is tested in three to four sequential

    trials at approximately 5 min intervals to avoid sensitization of the response. One of the advantages of this method

    versus the tail-flick assay is that both paws can be tested. This important control has proven a useful behavioral

    assessment in models of unilateral inflammation or nerve injury, the contralateral paw serving as control for the injured

    paw. In addition, in this assay, rats are confined in plastic chambers but not manually restrained as in the tail-flick assay

    or in the immersion tests (see below), decreasing the stress level of the test subjects. This method also uses a heated

    (30 1C) glass test surface to prevent paw cooling and to minimize sensitization artifacts.

    Acute thermal pain can also be evaluated using a fixed temperature (4550 1C) water bath and assessment of latency

    to withdraw of a hind limb or tail from the hot water. One of the advantages of this method is that the water bath can be

    set at various temperatures and it can be less sensitive to environmental conditions. However, this assay requires

    handling of the animals when testing for nociceptive behavior, making this measure highly dependent on experimenter

    experience/comfort handling/restraining animals by hand.

    This method can also be used to test for reactivity to cold, using a 4 or 10 1C water bath and recording latency to

    withdraw as an index of pain. Another method uses a cold plate cooled by cold water circulating under it. Latency to

    nociceptive behavior or duration of guarding behavior can be recorded. As for the hot plate assay, the cold plate test has

    the advantage of not requiring animal restraint. However, depending on the position of the animal paw on the plate (or

    just above it), the cold stimulation can be very variable. Another widely used method is application of a drop of cold

    acetone on the plantar skin of animals resting on an elevated mesh floor. Acetone produces a distinct cooling sensation

    as it evaporates. Normal rats will not respond to this stimulus or with a very small response (in amplitude and duration)

    while nerve-injured rats will almost always respond with an exaggerated response.

    6.14.3.1.2 Acute mechanical painModels have been developed to interrogate acute mechanical pain/sensitivity, using various means of stimulating the

    paw or the tail of a mouse or a rat. A common method for the assessment of acute mechanical pain is determination of

    withdrawal thresholds to paw/tail pressure using the Randall Selitto test.28 This apparatus allows for the application of a

    steadily increasing pressure to the dorsal surface of the hind paw/tail of a rat via a dome-shaped plastic tip. The

    threshold (in g) for either paw/tail withdrawal or vocalization is recorded. Usually, two or three measurements are

    conducted on each paw or tail. This apparatus was designed originally for measuring mechanical sensitivity of inflamed

    paws and its use in normal noninflamed paws can produce great variability in the response, depending on the location of

    the stylus (soft tissue between the metatarsal/bone/joint). It is worth noting that training helps generate a more stable

    response with this assay.

    Another approach for assessment of acute mechanical pain is to use a pinprick, applying painful pressure to the

    plantar surface of the hind paw. This is similar to the pricking pain test done during the neurological examination in

    patients. The behavior can be measured by the duration of paw lifting following the pinprick application or recorded as

    a frequency of withdrawal (percentage of response to the pinprick in 10 trials).

    Finally, mechanical hypersensitivity can also be tested with von Frey monofilaments. These are a series of hairs/

    nylon monofilaments of various thicknesses that exert various degrees of force when applied to the planter surface of

    the hind paw. Responses can be quantified as percentage response or duration of response to a given monofilament

    force applied several times, or mechanical threshold can be determined using the updown method. 29

    6.14.3.1.3 Acute chemical pain

    Usually, when studying acute chemical pain, behaviors such as flinching, biting, or licking the injected paw are

    recorded at various time points following the injection of a chemical irritant (capsaicin, formalin, PGE2, mustard oil,

    ab-methylene ATP). The duration of the nociceptive behavior as well as the number of behaviors can be quantified and

    analyzed. Two models are mostly used to study acute chemical pain: nocifensive behaviors following injection of

    332 Acute and Neuropathic Pain

  • 7/30/2019 Acute and Neurophatic Pain

    7/23

    capsaicin or formalin into the hind paw.30 Doses of capsaicin can vary from 1 to 10 mg per 10 mL injected to the dorsal

    surface of the rat hind paw. The injection of capsaicin is immediately followed by an intense period of nocifensive

    behaviors that are usually recorded for 5 min following capsaicin injection. Following formalin injection (usually 5% per

    50mL) into the dorsal or sometimes plantar surface of the rat hind paw, a biphasic behavioral response can be observed.

    Phase I of the formalin response is defined as the period of time immediately following injection of formalin until

    10 min after the formalin injection and corresponds to acute thermal pain by direct activation of nociceptors by

    formalin. Following a quiet period of little or no nocifensive behavior, the second phase of the formalin response can be

    observed (2060 min post formalin injection) that corresponds to a more persistent inflammatory state.

    6.14.3.2 Models of Nociceptive Pain

    Models of nociceptive pain are defined as models of pain following tissue injury induced by trauma, surgery,

    inflammation, and cancer. As stated above, spontaneous pain in these models is difficult to measure. However, evoked

    pain behaviors have been well characterized and can be induced by the methods described above in the acute pain

    section. The focus of this section will be on models of nociceptive pain, mimicking as closely as possible rheumatoid

    arthritis and osteoarthritis clinical conditions since they have been the most studied and widely used. Models of

    postoperative pain or cancer pain will not be described, as they are recent and still under validation.

    6.14.3.2.1 Adjuvant-induced arthritis

    Experimental arthritis is generated by an intravenous injection of complete Freunds adjuvant (CFA) at the base of the

    tail. The development of the joint inflammation is progressive and dramatic, leading to a multijoint arthritis with

    dramatic swelling and permanent joint tissue destruction.31 In this model, it is clear that the animals are in chronic

    pain, all their joints are swollen, they have decreased appetite, they limp, and have lower threshold for limb withdrawal

    or vocalization to paw pressure/joint manipulation. This model is rarely used today as the polyarthritic rat has significant

    systemic disease with abnormal hunchback posture and piloerection.

    6.14.3.2.2 Unilateral inflammation

    To further study inflammatory pain, various models have been developed to induce a localized inflammatory reaction by

    injecting various substances, e.g., formalin, carrageenan, or CFA into the paw or the joint. Following the initial injection,

    pain can be measured minutes to days later, at the site of inflammation or away from the primary site of injury. Usually,the inflamed paw/joint becomes very sensitive to both thermal and mechanical stimuli while the contralateral paw

    remains normal. Sometimes, secondary mechanical hypersensitivity can also develop on the contralateral side as

    observed 2 weeks following carrageenan injection into the knee joint when testing on the contralateral paw. These

    models of more localized inflammation/inflammatory pain have been widely used in pain research to test the effects of

    potential analgesic compounds but also in electrophysiological and gene expression studies to determine the plastic

    changes that initiate/maintain chronic inflammatory pain.

    6.14.3.2.3 Models of osteoarthritic pain

    More recently, models have been developed to mimic osteoarthritic (OA) pain observed in the clinic. Contrary to

    rheumatoid arthritis (RA) and the models of inflammatory pain, OA in the clinic and in animal models is not associated

    with a large amount of inflammation. In addition, to mimic more closely the clinical situation, pain evaluation in OApain models relies on functional measures such as weight bearing or grip force of the affected limb rather than

    evaluation of withdrawal latencies to thermal or mechanical stimuli. Two models have been widely used, intraarticular

    administration of sodium monoiodoacetate (MIA) into the knee and partial meniscectomy. 32 Contrary to what is

    observed in the polyarthritic rat, no changes in body weight were observed over a 4-week period after either iodoacetate

    injection or partial medial meniscectomy. In addition, the general health of the animals is good with no signs of

    spontaneous nociceptive behavior, impaired locomotion, or distress. Furthermore, both iodoacetate injection and partial

    medial meniscectomy in the knee joint of the rat induced histological changes and pain-related behaviors characteristic

    of clinical OA. Although the behavioral changes and histology both worsened over time, the majority of the pain

    responses were apparent within one week of surgery or iodoacetate injection. It is important to note that the pain

    behaviors are less pronounced in the surgery model than in the MIA model and that these findings agree with the

    clinical situation. Indeed, magnetic resonance imaging (MRI) studies have shown that although meniscal lesions in

    humans are common, they are also rarely associated with pain.

    Acute and Neuropathic Pain 333

  • 7/30/2019 Acute and Neurophatic Pain

    8/23

    6.14.3.3 Models of Neuropathic Pain

    6.14.3.3.1 Direct trauma to nerves

    To mimic nerve injury observed in the clinic, a number of different animals models have been developed. One of the most

    studied models is the L5L6 spinal nerve ligation (SNL, Chung model) (Figure 4) model.33 In this model, following

    sterilization procedures, a 1.5 cm incision is made dorsal to the lumbosacral plexus, the paraspinal muscles are separated from

    the spinous processes, the L5 and L6 spinal nerves are isolated, and tightly ligated with 30 silk thread. Usually the animals

    are allowed to recover from surgery for 7 days before being tested for mechanical allodynia using von Frey monofilaments(updown method or percentage response to 10 applications of innocuous or noxious von Frey monofilament). While the

    spinal nerve injured rats also develop cold allodynia and thermal hyperalgesia, they have a greater degree of mechanical

    allodynia and most pharmacological studies with these animals have involved mechanical allodynia endpoints.

    Another widely used model of direct nerve injury is a partial nerve ligation model (PNL) (Figure 4).33 The sciatic

    nerve is exposed unilaterally, just distal to the descendence of the posterior biceps semitendinosus nerve from the

    sciatic. The dorsal 1/31/2 of the nerve thickness is then tightly ligated with an 80 silk suture. Following injury, these

    animals develop guarding behavior of the injured hind limb suggesting the possibility of spontaneous pain. In addition,

    the animals develop mechanical allodynia as well as thermal hyperalgesia and bilateral mechanical hyperalgesia.

    6.14.3.3.2 Inflammation/neuritis/nerve compression

    Neuropathic pain can also result from inflammation around peripheral nerves and peripheral nerve compression. Two

    preclinical models have been developed to attempt to mimic this phenomenon. The first model is the chronic

    constriction injury (CCI) (Bennett model) (Figure 4) of the sciatic nerve model.33 In this model, a 1.5 cm incision is

    made 0.5 cm below the pelvis. The biceps femoris and the gluteus superficialis are separated and the sciatic nerve

    exposed, isolated, and four loose ligatures (50 chromic catgut) with 1 mm spacing are placed around it. CCI animals

    develop mechanical allodynia, cold allodynia, and thermal hyperalgesia. When compared to the SNL injured animals,

    CCI animals do develop thermal hyperalgesia and cold allodynia to a greater extent.

    The second model, developed more recently, is the SIN model or zymosan-induced sciatic inflammatory neuritis.34

    In this model, a chronic indwelling perisciatic catheter is used to inject zymosan around the sciatic nerve. After aseptic

    exposure of the sciatic nerve at midthigh level, the gelfoam is threaded around the nerve so as to minimize nerve

    displacement. Suturing and insertion of a sterile dummy injection tube during implantation maintained catheter

    patency and ensured replicable drug delivery close to the nerve. After anchoring to the muscle, the external end is

    tunneled subcutaneously to exit 1 cm rostral to the tail base. After removal of the dummy injector, the external end ofthe silastic tube is protected. Usually, catheter placement can be verified at sacrifice by visual inspection. The catheter

    Sural

    PNL

    CCISciatic

    SNL

    Peroneal

    Tibial

    DRG

    L4

    L4L5

    L6 Spinal cord

    Figure 4 Animal models of neuropathic pain. This schematic illustrates the three main rodent models of neuropathic pain

    associated with direct nerve injury; the L5L6 spinal nerve ligation (SNL) model (Chung model), the chronic constriction injury

    (CCI) of the sciatic nerve model (Bennett model), and the partial nerve ligation (PNL) model (Seltzer model).

    334 Acute and Neuropathic Pain

  • 7/30/2019 Acute and Neurophatic Pain

    9/23

    is used for a single injection 45 days after surgery conducted in freely moving rats. In this model, perisciatic zymosan

    injection induces unilateral mechanical allodynia at low dose and bilateral mechanical allodynia at high dose.

    Interestingly, the same high dose injected into gelfoam in neighboring muscles does not induce mechanical allodynia,

    suggesting that immune activation must occur in close proximity to peripheral nerves to create allodynia and that

    zymosan spread to systemic circulation cannot explain allodynia created by perisciatic zymosan. Interestingly, no

    thermal hyperalgesia is observed in this model.

    6.14.3.3.3 Diabetes

    Another major cause of neuropathic pain in the clinic is neuropathic pain observed in diabetic patients. In rodents, this

    is mimicked by streptozotocin (STZ) injection to induce diabetes and subsequent neuropathic pain symptoms. 33

    Usually, diabetes is induced by a single injection of STZ (75 mg kg1 intraperitoneal). Diabetes is confirmed by testing

    for blood glucose levels. Not all animals show signs of neuropathic pain immediately following STZ administration.

    Generally it takes usually between 4 and 8 weeks to observed neuropathic pain symptoms, mostly mechanical allodynia

    assessment with von Frey monofilaments, in a group of streptozotocin-treated rats.

    6.14.3.3.4 Chemotherapy-induced neuropathic pain (vincristine/paclitaxel/platine)

    The last type of neuropathic pain models are chemotherapy-induced neuropathic pain models. Cancer-related pain is

    a significant clinical problem that will likely increase in its extent as the average lifespan continues to rise and

    cancer therapies continue to improve. The two main sources of cancer-related pain are that from the malignancy itselfand from the treatments utilized to alleviate the cancer (surgery, radiation, and chemotherapy). Peripheral neuro-

    pathy and subsequent neuropathic pain related to chemotherapeutic treatment can be dose limiting, and the pain is

    often resistant to standard analgesics. To date, no one drug or drug class is considered to be both a safe and effective

    analgesic in the treatment of chemotherapy-induced pain, and three preclinical models of chemotherapy-induced

    neuropathic pain have been recently developed to further our understanding of the pathophysiology of such

    neuropathic pain states. Chemotherapy-induced neuropathic pain can be induced by the injection of either vincristine,

    platine, or paclitaxel.33 Depending on the experimental protocol, they can be injected as a bolus, for several days

    or weeks or as a continuous intravenous infusion using osmotic pump. Interestingly, as observed in the clinic,

    thermal hyperalgesia is not observed in these animals. However, both mechanical allodynia and cold allodynia are

    observed.

    The differential efficacy of analgesic medications for different types of pain that is seen in the clinic is also observed

    in animal pain models. For example, while opioid analgesics like morphine (Figure 5) are potent and efficacious in allanimal pain models, anti-inflammatory agents such as ibuprofen and celecoxib (Figure 5) are most potent and effective

    in animal models associated with inflammation, and anti-epileptics like lamotrigine and gabapentin ( Figure 6) are most

    potent and efficacious in animal models of neuropathic pain (Table 1). As preclinical models of the various forms of pain

    appear to have selective and differential predictive validity for efficacy in the clinical setting, they should be useful in

    determining if new chemical entities (NCEs) with a novel molecular mechanism have the promise to be broad-

    spectrum analgesics.

    6.14.4 Clinical Trial Issues

    Traditionally, the assessment of novel analgesics has been based on methods and models based on the clinical utility of

    opioid analgesics.14

    Many of the endpoints measured involve the use of self-report methodologies including theclassical visual analog scale (VAS) with which patients rate their pain from a score of 0 (no noticeable pain) to 10 (worst

    pain imaginable). For the specific assessment of neuropathic pain, clinical studies have used tools like the McGill Pain

    Questionnaire, Neuropathic Pain Scale, and the Neuropathic Pain Symptoms Inventory. While the use of many of these

    analgesic endpoints has been validated in the clinical setting, the use of specific combinations of these scales may be

    useful to enhance the sensitivity of clinical outcomes for new analgesic compounds.

    Clinical trial designs often employ parallel placebo-controlled and randomized withdrawal types of experimental

    manipulations.14,35 The majority of these designs have been well validated using opioids; however, the relative utility in

    assessing novel analgesics that target specific aspects of chronic pain (e.g., neuropathic allodynia) await further evaluation

    (e.g., non-NSAID (nonsteroidal anti-inflammatory drug) mediated analgesia in the third molar extraction model).

    A number of nociceptive tests have also been used in experimental clinical trials including acute heat sensitivity,

    topical and intradermal capsaicin, heat/capsaicin combinations, and quantitative sensory testing using both mechanical

    and thermal stimuli.36,37

    Some of these tests can also be coupled to other functional readouts such as functional MRI

    Acute and Neuropathic Pain 335

  • 7/30/2019 Acute and Neurophatic Pain

    10/23

    (fMRI) and nerve conduction velocity assessments to further enhance measurements of drug action.38 For example, in

    assessing the acute antinociceptive effects of remifentanyl, the apparent sensitivity of fMRI analysis of human cortical

    oxygen utilization was significantly greater than patients subjective ratings using traditional VAS scales. The use of

    fMRI coupled with experimental pain models like the capsaicin-evoked secondary hyperalgesia model may provide a

    reliable early assessment of novel analgesic efficacy. Many of these techniques have received clinical validation using

    opioids and are now being used to characterize mechanistically novel therapeutics.

    NH2 CO2H NH2 CO2H

    CH3H3C

    N

    NH2O NCH3

    CH3

    N

    N N

    Cl Cl

    NH2

    NH2

    CH3NH

    S

    O

    Gabapentin Carbamazepine Amitriptyline

    Lamotrigine Duloxetine

    Pregabalin

    Figure 6 Analgesic adjuvant agents.

    Morphine Oxycodone Fentanyl Tramadol

    O

    NCH3

    H

    HO

    HO

    O

    NCH3

    OH

    HO

    O

    N

    N

    H3C

    O CH2N(CH3)2

    HO

    H

    CH3O

    NN

    F

    F

    F

    H3C

    SO2NH2

    O

    H3CO2S

    O

    Celecoxib Rofecoxib

    H3C

    CH3COOH

    CH3N

    COOH

    CH3

    CH3OCl

    O

    Ibuprofen Indomethacin

    O CH3

    O

    COOH

    Aspirin

    Figure 5 Opioid analgesics, nonsteroidal anti-inflammatory analgesics, and cyclooxygenase-2 (COX-2) inhibitors.

    336 Acute and Neuropathic Pain

  • 7/30/2019 Acute and Neurophatic Pain

    11/23

    6.14.4.1 Translational Medicine in Pain

    A major issue in all aspects of drug discovery is the extent of predictivity of the preclinical animal models used to

    characterize NCEs to the clinical situation. Of all areas of research, based on validation with opioids and nonsteroidal

    anti-inflammatory drugs (NSAIDs), pain models were always considered highly robust. However, the recent failure of

    multiple NCEs targeted for the neurokinin-1 (NK-1) or substance P receptor has raised serious concerns about the

    translation of preclinical analgesic data to analgesic efficacy in patients.39,40 Structurally diverse NK-1 antagonists from

    several companies were evaluated clinically for acute pain (third molar extraction, migraine, or diabetic neuropathy).40

    While these NCEs attenuated nociceptive responses sensitized by either inflammation or nerve damage, they

    exhibited little effect on baseline (acute) nociception in preclinical models of pain. However, these same NCEs failed

    to demonstrate significant analgesic efficacy in early clinical trials. This situation has led to the re-evaluation of both

    the predictability of the preclinical animal models as well as the utility of clinical trial designs that were customized to

    assess analgesic by opiate and/or NSAID mechanisms. Is should also be noted that with the exception of the formalinmodel described above, animal pain models largely depend on stimulus-evoked pain behaviors as experimental

    endpoints. Consequently, assessments of ongoing nociception that are the foundation of most clinical studies are not

    modeled very well preclinically. It remains unclear whether the disconnect between the preclinical and clinical data for

    NK-1 antagonists reflects species-dependent roles of substance P in chronic pain or an imprecision in relating the

    nociceptive state of experimental animals to that of humans. Clearly, additional research is needed to adequately

    resolve these translational research issues for NK-1 antagonists. This issue highlights the practical need for the

    development of accurate and cost-effective translational medicine approaches to assess analgesic efficacy such as the

    coupling of fMRI studies with pain modeling in clinical studies described above.38

    6.14.5 Current Treatments

    Currently available analgesic agents can be broadly categorized as nonopioid analgesics (acetaminophen and NSAIDs),opioid analgesics (morphine and fentanyl), and adjuvant analgesics or coanalgesics (e.g., antiepileptics, anesthetics, and

    antidepressants). Nonopioid analgesics are mostly used to relieve mild to moderate nociceptive pain, adjuvant

    analgesics are used to relieve neuropathic pain, and opioid analgesics are used to treat severe pain of all origins,

    depending on the dose prescribed. Based on a number needed to treat analysis, a variety of opioid medications

    including fentanyl and oxycodone produce equivalent analgesic efficacy in neuropathic pain as compared to gabapentin,

    tramadol, and analgesic adjuvants like tricyclic antidepressants and anticonvulsants.41 The primary adverse event

    associated with chronic opioid therapy is decreased gastrointestinal (GI) motility. Respiratory depression and opioid

    dependence, which are routinely cited as a major issue in the use of opioids for pain therapy, are significantly less

    prevalent in chronic pain patients. This finding has led to ethical concerns related to patients not being given sufficient

    pain medication due to legal issues with drug scheduling.35,42

    While currently available analgesics have therapeutic utility in different pain states, all suffer from drawbacks in

    clinical use. The opioids can produce tolerance and dependence, constipation, respiratory depression, and sedation.

    Table 1 Effects of clinically used analgesics in preclinical models of acute, nociceptive, and neuropathic pain; relative

    analgesic efficacy of clinically useful analgesics in experimental pain models in the absence of psychomotor side effects. Data

    derived from both in house and literature values (see text)

    Acute pain Nociceptive pain Neuropathic pain

    T C F Car CFA OA RA SNL CCI Vinc

    Morphine 2 2 3 4 5 4 5 2 3 3

    Ibuprofen 0 1 2 5 5 2 NT 2 3 0

    Celecoxib 0 1 2 5 5 5 1 2 3 0

    Gabapentin 0 1 3 2 3 2 NT 5 5 3

    Lamotrigine 2 2 3 2 3 2 NT 5 4 3

    Scale: 0, no effect; 1, o20% effect; 2, 2040% effect; 3, 4060% effect; 4, 6080% effect; 5, 80100% effect, NT, Not

    tested. Abbreviations: T, tail flick; C, abdominal constriction; F, formalin; Car, carrageenan; CFA, complete Freunds

    adjuvant; OA, osteoarthritis; RA, rheumatoid arthritis; SNL, spinal nerve ligation; CCI, chronic constriction injury; Vinc,

    vincristine.

    Acute and Neuropathic Pain 337

  • 7/30/2019 Acute and Neurophatic Pain

    12/23

    The NSAIDs are associated with GI side effects and increased bleeding time, and do not effectively ameliorate severe

    pain. It has been estimated conservatively that 16 500 NSAID-related deaths occur among patients with rheumatoid

    arthritis or osteoarthritis every year in the USA.43 The development of selective COX-2 inhibitors (e.g., celecoxib and

    rofecoxib (Figure 5)) offered NSAID-like analgesia with the potential for diminished GI side effects. However, in light

    of the withdrawal of rofecoxib from the market, the cardiovascular safety of this class of analgesics is questionable. 44

    Adjuvant agents, e.g., gabapentin, lamotrigine, and amitryptiline, whose mechanism of action may be primarily

    mediated via a nonselective block of sodium channels, are associated with CNS and cardiovascular side effects.

    Currently available analgesics also have limited utility in the treatment of neuropathic pain. The anticonvulsant

    adjuvant gabapentin (Figure 6), which also has a poorly defined mechanism of action, has demonstrated clinical utility

    in the treatment of some forms of neuropathic pain.45 Thus, there is a significant unmet medical need for safer and

    more effective analgesic agents.

    6.14.5.1 Opioids

    Opioid analgesics produce their effects by binding and activating the opioid receptor subtypes (e.g., m, d, k receptors)

    in the CNS.6 The cloning and characterization of the major opioid receptor subtypes (m, OP1; d, OP2; and k, OP3) has

    stimulated significant basic and clinical research to discover new opioids with improved target selectivity, safety, and

    efficacy. Moreover, each of these opioid subtypes has been further subdivided into putative subtypes, and an orphan

    member of this family, ORL-1 (OP4), has also been described.

    46

    Given the anatomical distribution of the opioid receptors in the CNS, opioid analgesics can block pain transmission

    from the periphery to the spinal cord by blocking neurotransmitter release from the primary afferent fibers and by

    directly decreasing activation of postsynaptic dorsal horn neurons. The broad spectrum analgesic efficacy of the opioids,

    like morphine, fentanyl, and oxycodone (Figure 5), coupled with the fact that these agents do not show analgesic

    ceiling effects makes opioid compounds the mainstay in the control of moderate to severe pain. 46 The analgesic actions

    of opioid drugs are mediated at multiple sites of action including primary sensory afferent neurons, the dorsal horn of

    the spinal cord, and sites within the brain such as the brainstem and midbrain.

    This multitude of opioid interactions also contributes to the side effects associated with opioid analgesic therapy

    including dependence, tolerance, immunosuppression, respiratory depression, and constipation.41 Opioid dose titration

    can be achieved to manage some nociceptive conditions; however, this strategy does not provide full efficacy in all

    chronic pain syndromes such as cancer and neuropathic pain.35,41 In addition, acting at higher brain centers, opioid

    analgesics can decrease pain transmission from the spinal cord to the brain, alter the limbic system, and increasedescending inhibitory pathways to modulate pain transmission at the spinal level. While nociceptive pain is generally

    more responsive to opioid analgesics than neuropathic pain, nearly all types of pain respond to the right dose of opioid

    analgesics. Opioid analgesics are usually recommended to treat moderate to severe pain that does not respond to

    nonopioid analgesics alone and are often prescribed in combination with nonopioid analgesics. They do play a key role

    in the treatment of acute pain (postoperative pain), breakthrough pain, cancer pain, and some types of chronic

    noncancer pain.35 However, the use of opioid analgesics is associated with side effects such as sedation, confusion,

    respiratory depression, constipation, nausea, and vomiting that can limit their utility. More recently, a number of

    controlled release or extended release formulations of opioids have been developed in an effort both to enhance the

    analgesic coverage of these medications and to reduce the severity of the adverse events.

    The discovery of the opioid receptors in 1973 and their subsequent cloning has conceptually provided the tools

    necessary to develop receptor subtype selective NCEs that may improved efficacy and/or reduced side effect liabi-

    lities that would make them more useful analgesic agents. However, since 1975, more than $3 billion has beenspent on research to find such improved NCEs but none has lived up to its preclinical promise in the clinical

    situation.

    6.14.5.2 Anti-Inflammatory Drugs

    6.14.5.2.1 Nonsteroidal anti-inflammatory drugs

    The primary mechanism of action of NSAIDs is the blockade of prostaglandin synthesis via inhibition of the cyclo-

    oxygenase enzymes (COX-1 and COX-2). NSAIDs like aspirin, diclofenac, and ketorolac (Figure 5) have anti-

    inflammatory, antipyretic, and analgesic effects, and their anti-inflammatory effects can indirectly relieve nociceptive

    pain by reducing inflammation and tissue swelling, although potency across these effects can be compound dependent.

    These characteristics make them particularly efficacious is nociceptive pain conditions associated with peripheral

    inflammation.

    338 Acute and Neuropathic Pain

  • 7/30/2019 Acute and Neurophatic Pain

    13/23

    6.14.5.2.2 Cyclooxygenase-2 inhibitors

    Recently, selective inhibitors of COX-2 (the COX enzyme isoform inducible by inflammation), e.g., celecoxib and

    rofecoxib (Figure 5), have demonstrated similar efficacy to classical NSAIDs with reportedly fewer GI side effects at

    therapeutic doses.47 However, this is controversial and these agents have also recently been associated with a

    significant increase in the risk of cardiovascular disease that has brought the entire class under intense scrutiny with

    rofecoxib having been withdrawn from the market.44,48 As stated above, NSAIDs relieve mild to moderate pain

    associated with trauma, surgery, cancer, and arthritis. They are especially effective for certain types of somatic pain such

    as muscle and joint pain, sprains, bone/dental pain (tooth extraction), inflammatory pain, osteoarthritic pain, and in

    combination with disease-modifying therapies in rheumatoid arthritic pain.49

    6.14.5.3 Analgesic Adjuvants

    Adjuvant analgesics have a primary indication other than pain but have demonstrated analgesic effects in particular

    pain conditions. Antiepileptic drugs such as gabapentin, amitriptyline, carbamazepine, and lamotrigine (Figure 6) are

    one type of adjuvant analgesic and are used primarily in the treatment of neuropathic pain. 45 While it is hypothesized

    that their analgesic effects are due to their ability to reduce membrane excitability and tone down a hyperexcitable

    nervous system, their exact mechanism(s) of action remains unclear and not all antiepileptic drugs are good analgesics.

    As a class, they are most efficacious at treating peripheral neuropathic pain, postherpetic neuralgia, trigeminal

    neuralgia, and diabetic neuropathic pain.50

    6.14.5.3.1 Amitriptyline

    Amitriptyline is a tricyclic compound that has been approved for treatment of major affective disorders (e.g.,

    depression) since the 1950s.51 The antidepressant actions of amitriptyline generally are associated with blockade of the

    uptake of serotonin and norepinephrine in the CNS; however, amitriptyline possesses a multiplicity of other distinct

    pharmacological activities, e.g., antagonist actions at histamine, muscarinic, a1-adrenergic, and serotonin receptors at

    namomolar concentrations and at a number of ion channels (e.g., sodium, calcium, and potassium) at micromolar

    concentrations.52,53 Amitriptyline also has micromolar affinity for blocking the uptake of the antinociceptive and anti-

    inflammatory purine adenosine.52 In addition, amitriptyline has peripheral anti-inflammatory/analgesic actions in

    several in vivo models that are associated with acute, local delivery of low concentrations of amitriptyline. Many of

    these pharmacological activities are likely to contribute to its analgesic and anti-inflammatory actions.

    6.14.5.3.2 Antiepileptics

    Recent data suggest that newer antiepileptic drugs such as gabapentin or lamotrigine (Figure 6) are better alternatives

    to older agents of this class. Lamotrigine has been shown to be effective in patients with trigeminal neuralgia, complex

    regional pain syndrome, and neuropathic pain associated with multiple sclerosis and HIV infection.54,55 In addition, it

    has been shown that gabapentin provides pain relief in diabetic neuropathic pain conditions and postherpetic neuralgia;

    it also has a more favorable side effect profile compared to other neuropathic pain agents.55 Open-label studies suggest

    that gabapentin also may be useful in the management of trigeminal neuralgia, central pain, phantom limb pain, and

    neuropathy associated with HIV infection. Common side effects of antiepileptic drugs as a class include sedation,

    mental clouding, dizziness, nausea, or unsteadiness, and patients need to start at low doses and go through a slow

    titration in order to diminish the risk of side effects.

    6.14.5.3.3 PregabalinPregabalin (Figure 6), an alkylated analog of GABA, is a more potent anticonvulsant than gabapentin and has shown

    enhanced analgesic potency as well.56 Pregabalin is clinically effective in pain associated with diabetic neuropathy and

    postherpetic neuralgia. The enhanced potency of pregabalin relative to gabapentin appears to be related to

    pharmacokinetic properties rather the mechanism of action. In this regard, the analgesic mechanism(s) for these

    anticonvulsant compounds has not been definitively determined. Recent data indicate that these GABA analogs bind

    the a2d subunit of voltage-gated calcium channels with high affinity.57 However, it should be noted that binding to the

    a2d subunit is not likely to fully account for the analgesic properties of gabapentin or pregabalin.58

    6.14.5.3.4 Antidepressants

    In addition to antiepileptic drugs, tricyclic antidepressant drugs like amitriptyline are also used as adjunct analgesics in

    the treatment of neuropathic pain.14,45 While other antidepressant medications such as selective serotonin reuptake

    inhibitors (SSRIs) have not proven to be particularly efficacious in treating neuropathic pain, some of the newer

    Acute and Neuropathic Pain 339

  • 7/30/2019 Acute and Neurophatic Pain

    14/23

    combination serotoninnorepinephrine reuptake inhibitors such as duloxetine and venlafaxine significantly attenuate

    neuropathic pain. Indeed, duloxetine (Figure 6) is the newest approved medications for the treatment of the pain

    associated with diabetic neuropathy.59

    Interestingly, in parallel to their preferred efficacy in the clinic, clinically used analgesics also have a differential

    analgesic profile in preclinical pain models (see Table 1). While opioid analgesics like morphine are potent and efficacious

    in all animal models of pain, including acute pain, pain associated with inflammation in which they are particularly potent,

    and pain associated with neuropathy, NSAIDs such as ibuprofen and celecoxib are most potent and effective in animal

    models associated with inflammation, with near to no efficacy in models of acute pain and neuropathic pain, and

    antiepileptics like lamotrigine and gabapentin are most potent and efficacious in animal models of neuropathic pain.

    6.14.6 Unmet Medical Needs

    Due to the relatively poor efficacy to tolerability ratio for opioid analgesics in treating neuropathic pain, this area

    represents a major unmet medical need. While gabapentin and other adjuvant analgesics have been reported to be

    clinically effective in treating neuropathic pain, the efficacy rates are relatively small and are often accompanied by side

    effects of sufficient magnitude to limit compliance.7 This situation is not surprising in that essentially all clinically used

    analgesic adjuvants were originally developed to treat other indications. Their clinical utility in treating pain has been

    based largely on clinical serendipity.

    In the case of inflammatory or nociceptive pain, opioids provide significant analgesic efficacy; however, long-termuse of these analgesics is limited by both opioid-mediated side effects including constipation, and regulatory concern of

    opioid dependence and abuse liability. NSAIDs also provide moderate pain relief in these pain states, but are associated

    with GI disturbances. The COX-2 inhibitors represent an analgesic advance due to their enhanced GI safety profile;

    however, the long-term cardiovascular safety of these agents is controversial.

    6.14.7 New Research Areas

    The identification clinically useful analgesic targeting nonopioid mechanisms has been challenging as demonstrated by

    the failure of several novel tachykinin NK-1 receptor antagonists in clinical trials.39 Other novel analgesic agents have

    either not been advanced or are used in only limited conditions due to mechanism related toxicities. For example, the

    analgesic efficacy of ziconotide, a selective neuronal calcium channel (N-type, Cav2.2) blocker (discussed in more detailbelow) is limited to intrathecal administration in order to minimize severe cardiovascular adverse effects. Additionally,

    the development of several classes of adenosine kinase inhibitors as analgesics was halted due to the occurrence of

    vascular microhemorrhages in brain.60 Despite these difficulties in translating advances in pain neurobiology into clinical

    useful analgesics, a number of novel analgesic mechanisms and compounds have been identified and validated in

    preclinical models. Some of these are described below.

    6.14.7.1 Neuronal Nicotinic Receptor Agonists

    Activation of neuronal nicotinic receptors (NNRs) represents a novel approach to pain management supported by the

    observation that epibatidine (isolated from Epipedobates tricolor) had significantly greater analgesic potency than

    morphine in assays of acute thermal pain.61,62 While the mechanism of action of epibatidine was unknown, it was

    subsequently found to be a picomolar agonists at NNRs. NNR agonists with higher affinity for the a4b2 subunit (thepredominant subtype in the CNS) relative to the a1b1dg nicotinic acetylcholine receptor subunit (located at the

    neuromuscular junction) had analgesic efficacy with a larger therapeutic window from severe side effects than did

    epibatidine.6367 These observations facilitated the discovery of ABT-594 (Figure 7), an a4b2-preferring NNR agonist

    that was synthesized independently of the identification of the mechanism of action of epibatidine. ABT-594 has

    broad-spectrum analgesic activity in both acute (hot plate, tail flick, formalin) inflammatory (CFA), and neuropathic

    pain models. Importantly, ABT-594 showed less potential for analgesic tolerance than morphine in animal models and

    did not produce pharmacologic dependence.6365

    6.14.7.2 Vanilloid Receptor Modulators

    The analgesic actions of topically applied capsaicin, the active ingredient in hot chillies, has been known for many years;

    however, the clinical utility of vanilloid-derived analgesics has been limited by the initial burning sensation these

    340 Acute and Neuropathic Pain

  • 7/30/2019 Acute and Neurophatic Pain

    15/23

    compounds elicit. The cloning and characterization of the capsaicin-sensitive vanilloid receptor (TRPV1) has greatly

    enhanced understanding of the mechanism by which vanilloids, acid, and heat may alter nociceptor sensitivity.68 TRPV1

    receptors are one member for a larger family of transient receptor potential (TRP) proteins, several member of which

    have been associated with sensory function including cold (TRPA1, TRPM8) and mechanical (TRPV3, TRPV4)

    sensitivity.68 These findings have also led to the discovery of several distinct classes of TRPV1 antagonists (Figure 8).69,70

    These compounds potently and selectively block capsaicin activation of TRPV1 in a competitive manner. Interestingly,

    these antagonists also effectively block TRPV1 activation by acid and heat, indicating that these agents may exert channel

    modulating activity as compared to direct channel activation. These latter effects may be species dependent with some

    capsaicin-competitive antagonists being ineffective in blocking acid activation of rat TRPV1 receptors. TRPV1

    antagonists have demonstrated analgesic efficacy in animal models of inflammatory pain, thus illustrating their potential

    to be clinically effective analgesics.

    6.14.7.3 Excitatory Amino Acid Receptor Antagonists

    The excitatory amino acid (EAA) glutamate functions as a primary excitatory neurotransmitter in the CNS, and

    activation of EAA-specific ionotropic and metabotropic receptor superfamilies in the spinal cord underlies the process

    of central sensitization involved in chronic pain.16 Activation of the heteromultimeric NMDA receptors (NR1/NR2B/

    NR2D subunits) expressed in spinal cord contributes to the expression of tactile and thermal hyperalgesia. A number

    of competitive and noncompetitive NMDA receptor antagonists including (7)-CPP, MK-801, ketamine,

    and dextromethorphan (Figure 9) block hyperalgesia in animal models and attenuate the process of central

    sensitization.71,72 A problematic issue associated with NMDA receptor antagonists is their psychotomimetic effects

    that include both dysphoria and cognitive impairment. This has led to the search for ligands that are selective for

    specific NMDA channel subunits (e.g., NR2B) that may alter nociceptive processing with an improved therapeutic

    window relative to previous NMDA antagonists. CP-101,606 (Figure 9), a selective NR2B antagonist, effectivelyreduced pain in low back pain and spinal cord injury patients.73 Adverse events associated with CP-101,606 treatment

    were dizziness, and hypoesthesia, but were tolerated by the patients.

    Memantine (Figure 9) is a low-affinity (NR1/NR2B IC50820 nM) noncompetitive NMDA antagonist that has

    analgesic efficacy in humans. In early clinical studies, memantine attenuated ongoing neuropathic pain symptoms in

    both diabetic and postherpetic neuralgia patients that memantine failed in Phase III pain indication trials.74 In addition

    to these agents, other NMDA antagonists are being investigated preclinically as potential analgesic. For example,

    MRZ-2/579 (Figure 9), a low-affinity noncompetitive NMDA antagonist, attenuates carrageenan-induced thermal

    hyperalgesia at doses that do not affect sensory-motor function.75 GV 196771A (Figure 9) modulates NMDA receptor

    function by blocking the glycine binding site of the NMDA receptor complex. Like memantine and dextromethorphan,

    GV 196771A produces antihyperalgesia in animal models at doses that do not elicit CNS side effects.76Additionally, GV

    196771A is only weakly active in cerebral stroke models suggesting differences in the physiological substrates of

    nociception and neuroprotection. NMDA receptor antagonists can provide opioid sparing effects and may prevent the

    ABT-594

    NO

    HN

    Cl

    Figure 7 ABT-594, a neuronal nicotinic receptor (NNR) agonist.

    AMG 9810

    O

    OHN

    O

    OHN

    O

    Cl

    N

    NH

    HN

    O

    F

    FF

    SB-366791 A-425619

    Figure 8 Novel nonvanilloid TRPV1 receptor antagonists.

    Acute and Neuropathic Pain 341

  • 7/30/2019 Acute and Neurophatic Pain

    16/23

    tolerance related to prolonged opioid use. As noted above, late stage clinical trials of a dextromethorphan/morphine

    combination (MorphiDex) are ongoing. Antagonists of the kainic acid subtype of the glutamate receptor include LY

    293558 (Figure 9) which was active in acute migraine.77

    6.14.7.4 Calcium Channel Modulators

    Modulation of N-type (Cav2.2) calcium channels has been shown to provide an avenue for development of novel

    analgesics, as exemplified by ziconotide, a 25 amino acid polycationic peptide originally isolated from the venom of a

    cone snail. Its delivery is limited to the epidural and intrathecal routes, as systemic administration has led to a risk

    for orthostatic hypotension.78 Ziconotide is approved for treating intractable cancer pain and chronic neuropathic

    pain. The clinical efficacy of ziconotide provides important validation of this novel analgesic mechanism. In addition

    to N-type calcium channels, T-type channels (Cav3.1 and Cav3.2) have also been implicated in nociceptive processing.

    T-type calcium channels are expressed on dorsal root ganglion neurons and intrathecal antisense treatment targeting

    the Cav3.2 subtype of T-type calcium channels effectively blocked all low-voltage calcium currents in dorsal root

    ganglion neurons and significantly attenuated both acute and inflammatory pain.79

    6.14.7.5 Cannabinoids

    Marijuana (cannabis) has been used to relieve pain for centuries.80 However, clinical evaluation of the major active

    cannabinoid, D9-tetrahydrocannabinol (D9-THC), has produced equivocal results in chronic cancer pain patients.

    Furthermore, the analgesic actions of D9-THC could not be clearly separated from the other well-described

    psychotropic actions ofD9-THC. Investigation of the pharmacological actions of the cannabinoids has been greatly

    aided by the recent discovery of specific cannabinoid receptor subtypes (CB1 and CB2), elucidation of their signaltransduction pathways, and the identification of putative endogenous ligands (e.g., anandamide).80 High densities of

    CB1 receptors are found in the CNS, while CB2 receptors are localized primarily to immune cells and peripheral nerve

    terminals. These advances in cannabinoid pharmacology suggest the possibility of identifying receptor subtype

    selective ligands.

    Cannabimimetics have been shown to produce antinociception in animal pain models via spinal and supraspinal

    actions on CB1 receptors, and by peripheral actions at CB2 receptors on sensory afferents and, indirectly, on immune

    cells. Recent compounds in preclinical development include agonists with improved oral bioavailability and/or

    enhanced receptor subtypes selectivity. CT-3 (Figure 10) is an orally active and nonselective analog of THC that dose-

    dependently reduces acute nociception in the rat. Recently, CT-3 has been tested in the clinic in a phase II trial

    in chronic neuropathic pain patients and the results suggested that CT-3 could be useful in treating this condition.80

    O-1057 (Figure 10) is a potent and moderately CB1 receptor selective analog of CT-3 that has improved water

    solubility and acute antinociceptive actions.67

    HU-308 (Figure 10) is a novel, highly CB2 receptor selective agonist

    CH3

    CH3

    H2N

    Memantine

    HN

    O

    OH

    N

    O

    Cl

    Cl

    NH

    H

    H

    COOHNN

    NNH

    GV 196771A LY 293558

    NH3+

    H3C

    H3CH3C

    CH3CH3

    MRZ-2/579

    N

    OH

    HO

    OH

    CP-101,606

    O

    NCH3

    H

    CH3O

    Dextromethorphan

    Figure 9 Antagonists for excitatory amino acid (glutamate) receptors.

    342 Acute and Neuropathic Pain

  • 7/30/2019 Acute and Neurophatic Pain

    17/23

    (Ki CB1410 mM, CB223 nM) that has antinociceptive effects in the persistent phase of the mouse formalin test, but

    was inactive in the acute phase of the formalin test.81 While no effects of HU-308 were observed on motor function,

    antinociceptive doses of the compound also reduced GI motility and blood pressure.82

    More recently, the potential analgesic profile of CB2 agonists has been extensively characterized in in vivo pain

    models in rodents. PRS-211375 (Figure 10) has been shown to be CB2 selective (CB2 Ki9 nM; CB1 Ki300 nM)

    and a full agonist in adenylyl cyclase and GTPgS binding assays with similar potencies as observed in binding assays.

    PRS-211375 has good CNS penetration and has shown efficacy in various animal models including formalin,

    neuropathic pain models, acute thermal pain, visceral pain, and CFA-induced arthritis.83 PRS-211375 recently

    completed phase I clinical trial. AM-1241 (Figure 10) is another selective CB2 agonist used in preclinical models toidentify the potential analgesic profile of CB2 agonists. This compound has been shown to decrease acute,

    inflammatory, and neuropathic pain, its effects mediated through the release of b-endorphin and effects on opioid

    receptors.84 GW405833 (Figure 10), another CB2 agonist, has also been shown to produce analgesia in animal models

    of chronic pain including postoperative pain. However, contrary to the results obtained with AM-1241, the analgesic

    effects of GW405833 are not mediated through activity at opioid receptors.85

    6.14.7.6 Sodium Channel Modulators

    The activation of voltage-gated sodium channels is necessary for the generation of neuronal action potentials. A feature

    common to the local anesthetics and most analgesic adjuvants (e.g., carbamazepine, lamotrigine, and amitriptyline) is

    their ability to block sodium channels and this property may underlie the clinical utility of these agents in reducingpain. However, all of these agents possess other pharmacologically relevant activities that results in limits their

    analgesic effectiveness in the clinic. Notably, these include CNS sedation and/or untoward cardiovascular effects.

    The cloning and characterization of several sensory nerve-specific sodium channel subtypes has raised interest in

    the possibility of developing subtype-specific inhibitors which might overcome the cardiovascular and proconvulsant

    liabilities of nonselective agents. The voltage-gated sodium channel gene family consists of multiple members, termed

    Nav1.1 through Nav1.9.86 At least six of these channels are found in the peripheral nervous system.87 Structurally, the

    family has a high overall degree of similarity (around 50% identity), with subfamilies being very closely related (up to

    90% identity). Susceptibility to blockade by natural toxins, particularly tetrodotoxin (TTX), has been typically used to

    classify sodium channel currents. Two TTX-resistant channels are present in the periphery: Nav1.8 (also called PN3 or

    SNS) and Nav1.9 (also called NaN). Nav1.8 is likely to be the more important sodium channel in regulating nociceptive

    signaling since in vivo antisense experiments targeting Nav1.9 did not reduce chronic neuropathic pain.88 Nav1.8

    immunoreactivity is increased in the carrageenan inflammatory pain model, and increased proximal to the site of nerve

    OR

    O

    CO2H

    R = H

    R =O

    N+O

    H

    OCH3

    H3CO

    CH2OH

    CT-3

    O-1057

    HU-308

    N

    OI

    NO2N

    CH3

    AM-1241

    O

    O

    OH

    O

    OO

    PRS-211375

    N

    O

    N

    O

    Cl Cl

    OGW405833

    Steriochemistry

    Figure 10 Agonists for cannabinoid receptors.

    Acute and Neuropathic Pain 343

  • 7/30/2019 Acute and Neurophatic Pain

    18/23

    injury in rats and humans. Antisense oligonucleotides against Nav1.8 prevented thermal hyperalgesia or mechanical

    allodynia from developing in animal models of neuropathic pain, and were also effective at reducing prostaglandin-

    induced hyperalgesia. Nav1.8 knockout mice demonstrated a diminished response to noxious mechanical stimuli and

    delayed inflammatory hyperalgesia. While the PN3/SNS subtype has been a major focus of research, other sodium

    channels may also be appealing targets for pharmaceutical intervention of pain.89,90 These data indicate that the

    development of antagonist that are selective for specific sodium channel subunits, like Nav1.8 may provide novel

    analgesic agents. The feasibility of this approach is likely to be difficult based on the poor selectivity of currently

    available sodium channel blockers, however, two NCEs have been recently described that show increased potency for

    tetrodotoxin-resistant sodium channels as compared to the typical analgesic adjuvants. Ralfinamide (NW-1029)

    (Figure 11) inhibits TTX-resistant currents in rat dorsal root ganglion neurons with an IC50 value of 10mM and dose-

    dependently reduced allodynia in neuropathic pain models.91 CDA-54 (Figure 11) is a potent (IC50 B200 nM)

    nonselective sodium channel blocker that reduces pain in inflammation pain models.92

    6.14.7.7 Purines

    6.14.7.7.1 P1 receptor agonists

    The systemic or spinal administration of adenosine (ADO) or ADO A1 receptor selective agonists has been shown to

    provide clinically effective analgesia.93 However, analgesic efficacy of systemically administered ADO in neuropathic

    pain patients has been variable.94

    In contrast, intrathecal injection of ADO or an A1 receptor agonist appears toconsistently reduce pathological pain in neuropathic subjects or in healthy volunteers given intradermal capsaicin to

    induce central sensitization.9597 Despite these promising results, their profound effects on cardiovascular function

    have hampered the development of ADO receptors agonists as analgesics.

    Inhibition of the primary metabolic enzyme, adenosine kinase (AK), which regulates ADO availability enhances the

    beneficial actions of adenosine at sites of tissue injury or trauma and AK inhibitors are potent antinociceptive and anti-

    inflammatory agents with an potentially improved therapeutic window over direct acting ADO receptor agonists.98

    ABT-702 (Figure 12), a non-nucleoside AK inhibitor, has both analgesic (e.g., effective in acute nociception) and

    antihyperalgesic activity (e.g., effective in blocking both hyperalgesia and allodynia).98 The analgesic actions of ABT-

    702 are blocked by selectivity ADO A1 receptor subtype antagonists and the locus of analgesic activity is in the spinal

    cord.98ABT-702 also shows significantly greater separation between its antihyperalgesic activity and its effects on CNS

    or cardiovascular function as compared to direct-acting ADO receptor agonists.

    6.14.7.7.2 P2 receptor antagonists

    The cloning and characterization of the family of ATP-sensitive ligand-gated ion channels (P2X receptors) provided

    mechanistic insights for the role of ATP as an extracellular signaling molecule.99 The P2X3 channel is localized

    O

    F HN

    NH2O

    Ralfinamide CDA-54

    O

    HNN

    OH2NO2S OCF3

    Figure 11 Novel voltage-gated sodium channel blockers.

    N

    N N

    NH2

    N

    Br

    N

    OABT-702

    Figure 12 ABT-702, a nonnucleoside adenosine kinase (AK) inhibitor.

    344 Acute and Neuropathic Pain

  • 7/30/2019 Acute and Neurophatic Pain

    19/23

    primarily to sensory neurons, suggesting a role in pain transmission. The P2X3 messenger RNA (mRNA) occurs only in

    the trigeminal, dorsal root, and nodose ganglia, and the receptor is selectively expressed in sensory C-fiber neurons that

    project to the periphery and spinal cord, and which are predominantly nociceptors. In addition, P2X3 receptors located

    presynaptically at the central terminals of primary afferent neurons may have a facilitatory role to enhance

    neurotransmission, leading to a further increase in pain sensation.100

    A role of ATP in pain transmission is consistent with the observed induction of pain by ATP upon application to

    human skin, and with reports that intradermal and intrathecal application of ATP and ATP analogs (e.g., ab-methylene

    ATP (abmetATP)) into the rat hind paw evokes acute nociceptive behavioral responses. Transgenic disruption of P2X3receptors in rodents via knockout, antisense, or short interfering RNA (siRNA) manipulations leads to decreased

    nociceptive sensitivity.101,102 A-317491 (Figure 13), a potent and selective antagonist of homomeric P2X3 and

    heteromeric P2X2/3 receptors that when given systemically and intrathecally dose-dependently reduces nociception in

    animal models of inflammatory and neuropathic pain indicating that blockade of spinal P2X3 receptors may provide

    broad-spectrum analgesic effects in chronic pain states.100

    P2Y2 receptors signal through protein kinase C (PKC) and, in turn, modulates the activation of TRPV1 receptors.103

    Intrathecal antisense studies have also shown that P2X4 receptors acting via a spinal neuralmicroglial interaction alter

    nociceptive processing in neuropathic rodents.104 More recently, P2X7 receptor knockout mice have been characterized

    and show reduced inflammation and neuropathic pain as compared to wild-type mice.105

    P2X7 receptors are notlocalized on small-diameter neurons in the periphery, but are found on gial cells in the dorsal root ganglion.106 Blockade

    of P2X7 receptors leads to inhibition of IL-1b release from macrophages and glial cells, while antagonists like A-740003

    are active in pain models.110 In addition, P2X7 receptors mediate the ability of ATP to stimulate glutamate release from

    glial cells, thus providing an additional mechanism for ATP-mediated fast neurotransmission.107

    6.14.7.8 Emerging Pain Targets

    As indicated above, a developing concept in pain research is the appreciation that neuroimmune interactions

    participate in nociceptive signaling in chronic pain states.25 This has greatly added to the potential list of candidate

    mechanisms that may offer an avenue for analgesic intervention. In addition to traditional mechanisms associated with

    neuronneuron communication, cytokines, chemokines, and inflammatory acute phase proteins are now known to

    contribute to nociceptive signaling. This collection of potential analgesic mechanisms is further complemented byfindings that extracellular acid may also play a neuromodulatory role in the transmission of pain signals. Members of the

    TRP channel family are sensitive to both endogenous lipids as well as acidic pH.68,69 The activation of specific acid-

    sensing ion channels (ASICs) also contributes to the encoding of noxious stimulation.108 The development of selective

    antagonists of ASIC3 antagonist A-317567 (Figure 14) provided the demonstration that this acid-sensing channel

    contributes to both inflammatory hyperalgesia and pain associated with skin incision.108

    As noted above, neurotrophic factors play important roles in the remodeling of the peripheral and central nervous

    systems in response to pain. Specifically, nerve growth factor (NGF) is a neurotrophin that is an important survival

    factor for sensory neurons.109 However, NGF also has pronociceptive actions, producing pain and enhancing

    hyperalgesia in both experimental animals and in human clinical studies. The recent development of small-molecule

    antagonists like ALE0540 and PD90780 (Figure 15) as well as anti-NGF antibodies has enabled studies demonstrating

    that these agents effectively reduce chronic arthritic pain, skin incision pain, and tactile allodynia following peripheral

    nerve injury.109

    A-317491

    N

    O

    HO

    O

    OHHO O

    O

    O

    N

    N N

    N

    N

    N

    OOMe

    OMe

    A-740003

    Figure 13 A-317491 and A-740003, selective P2X3 and P2X7 ligand-gated ion channel antagonists, respectively.

    Acute and Neuropathic Pain 345

  • 7/30/2019 Acute and Neurophatic Pain

    20/23

    6.14.8 Conclusions

    The adverse physiological, psychological, and economic effects of inadequate pain management have become

    increasingly recognized in recent years. This has been accompanied by a growing awareness on the part of patients and

    caregivers that pain need not be tolerated, and an increased emphasis amongst physicians on the proactive treatment of

    pain. The unmet need for new analgesics remains substantial. Recent advances in the neurobiology of pain, together

    with the development of new preclinical and clinical pain paradigms, have revealed new opportunities for the

    development of analgesics, and raised the exciting possibility of entirely novel classes of analgesics in the future.

    References

    1. Merskey, H.; Bugduk, N. Classification of Chronic Pain, 2nd ed.; IASP Press: Seattle, WA, 1994.

    2. Harden, N.; Cohen, M. J. Pain Sympt. Mgmt. 2003, 25, S12S17.

    3. Elliott, A. M.; Smith, B. H.; Penny, K. I.; Smith, W. C.; Chambers, W. A. Lancet 1999, 354, 12481252.

    4. Ericksen, J.; Jensen, M. K.; Sjgren, P.; Ekholm, O.; Rasmussen, N. K. Pain 2003, 106, 221228.

    5. American living with pain survey; American Chronic Pain Association, Roper Public Affairs and Media: Rocklin, CA, 2004.

    6. Millan, M. J. Prog. Neurobiol. 1999, 57, 1164.

    7. Scholz, J.; Woolf, C. J. Nat. Neurosci. 2002, 5, 10621067.

    8. Cervero, F.; Laird, J. M. Pain 1996, 68, 1323.

    9. Woolf, C. J.; Costigan, M. Proc. Natl. Acad. Sci. USA 1999, 96, 77237730.

    10. Coda, B. A.; Bonica, J. J. In Bonicas Management of Pain, 3rd ed.; Lippincott: Baltimore, MD, 2001, pp 222240.

    11. Byers, M.; Bonica, J. J. In Bonicas Management of Pain, 3rd ed.; Lippincott: Baltimore, MD, 2001, pp 2672.

    12. Meyer, R. A.; Campbell, J. N.; Raja, S. N. Advances in Pain Research and Therapy; Raven: New York, 1985; Vol. 9, pp 5371.13. Abbott, J. D.; Moreland, L. W. Exp. Opin. Invest. Drugs 2004, 13, 10071018.

    14. Dworkin, R. H.; Backonja, M.; Rowbotham, M. C.; Allen, R. R.; Argoff, C. R.; Bennett, G. J.; Bushnell, M. C.; Farrar, J. T.; Galer, B. S.;

    Haythornthwaite, J. A. Arch. Neurol. 2003, 60, 15241534.

    15. Smith, P. A. Drug News Perspect. 2004, 17, 517.

    16. Woolf, C. J.; Mannion, R. J. Lancet 1999, 353, 19591964.

    17. Woolf, C.