Skip to main content Accessibility help
×
Hostname: page-component-68945f75b7-qvshk Total loading time: 0 Render date: 2024-08-05T18:40:54.881Z Has data issue: false hasContentIssue false

26 - Neuropathic pain

from SECTION IX - DIFFICULT PAIN PROBLEMS

Published online by Cambridge University Press:  06 July 2010

RICARDO A. CRUCIANI
Affiliation:
Beth Israel Medical Center
E. ALESSANDRA STRADA
Affiliation:
Beth Israel Medical Center
HELENA KNOTKOVA
Affiliation:
Beth Israel Medical Center
Eduardo D. Bruera
Affiliation:
University of Texas, Houston
Russell K. Portenoy
Affiliation:
Albert Einstein College of Medicine, New York
Get access

Summary

Introduction

Effective pain management in patients with cancer is an essential component of care during active treatment, as well as in palliative and end-of-life care. Chronic pain occurs frequently in patients with cancer, with an estimated prevalence that ranges from 40% to 60%. Pain may be caused by direct tumor infiltration of neural structures or by treatment sequelae. Although the prevalence of pain in the outpatient setting is 20%–40%, it may rise to 80% for patients at the end of life. However, several prospective studies suggest adequate pain control could be achieved in 90% of patients by following the World Health Organization guidelines.

Because pain is ultimately a perceptual construct within the central nervous system (CNS), a patient's experience of pain is a result of complex somatosensory processing. Developing understanding of the physiological and biochemical processes involved in pain, combined with an appreciation of the cognitive, emotional, and cultural framework of the patient offers the best opportunity to provide treatment of underlying mechanisms and relief of suffering.

Even though most patients initially report somatic or visceral pain, they also frequently experience painful neuropathic components. Management of neuropathic pain presents a number of challenges, because it is often more resistant to conventional analgesic approaches. Mixed pain problems, in which neuropathic pain is combined with elements of somatic or visceral pain, are also common. Neuropathic pain may also signal progressive and often incurable disease.

Type
Chapter
Information
Cancer Pain
Assessment and Management
, pp. 478 - 505
Publisher: Cambridge University Press
Print publication year: 2009

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Caraceni, A, Portenoy, RK. An international survey of cancer pain characteristics and syndromes. IASP Task Force on Cancer Pain. International Association for the Study of Pain. Pain 82:263–74, 1999.CrossRefGoogle Scholar
Cleeland, CS, Gonin, R, Hatfield, R, et al. Pain and its treatment in outpatients with metastatic cancer. N Engl J Med 330:592–6, 1994.CrossRefGoogle ScholarPubMed
Grond, S, Radbruch, L, Meuser, T, et al. Assessment and treatment of neuropathic cancer pain following WHO guidelines. Pain 79:15–20, 1999.CrossRefGoogle ScholarPubMed
Anderson, KO, Syrjala, KL, Cleeland, CS. How to assess cancer pain. In: Turk, DC, Melzack, R, eds. Handbook of pain assessment. New York, London: The Guilford Press, 2001, pp 579–600.Google Scholar
Katz, N. Neuropathic pain in cancer and AIDS. Clin J Pain 2000, 16:S41–8.CrossRefGoogle ScholarPubMed
Boucher, TJ, Okuse, K, Bennett, DL, et al. Potent analgesic effects of GDNF in neuropathic pain states. Science 2000, 290:124.CrossRefGoogle ScholarPubMed
Asbury, AK, Fields, HL. Pain due to peripheral nerve damage: a hypothesis. Neurology 34:1587–90, 1984.CrossRefGoogle ScholarPubMed
Elliott, KJ. Taxonomy and mechanisms of neuropathic pain. Semin Neurol 14:195–205, 1994.CrossRefGoogle ScholarPubMed
Chen, R, Cohen, LG, Hallett, M. Nervous system reorganization following injury. Neuroscience 111:761–73, 2002.CrossRefGoogle ScholarPubMed
Empl, M, Renaud, S, Erne, B, et al. TNF-alpha expression in painful and nonpainful neuropathies. Neurology 56:1371–7, 2001.CrossRefGoogle ScholarPubMed
Dworkin, RH, Backonja, M, Rowbotham, MC et al. Advances in neuropathic pain: diagnosis, mechanisms, and treatment recommendations. Arch Neurol 60:1524–34, 2003.CrossRefGoogle ScholarPubMed
Jin, SX, Zhuang, ZY, Woolf, CJ, et al. p38 mitogen-activated protein kinase is activated after a spinal nerve ligation in spinal cord microglia and dorsal root ganglion neurons and contributes to the generation of neuropathic pain. J Neurosci 23:4017–22, 2003.CrossRefGoogle ScholarPubMed
Bennett, GJ. Neuropathic pain. In: Wall, PD, Melzack, R, eds. Textbook of pain. Edinburgh: Churchill Livingstone, 1994, pp 201–24.Google Scholar
Elbert, T, Flor, H, Birbaumer, N, et al. Extensive reorganization of the somatosensory cortex in adult humans after nervous system injury. Neuroreport 5:2593–7, 1994.CrossRefGoogle ScholarPubMed
McGaraughty, S, Wismer, CT, Zhu, CZ, et al. Effects of A-317491, a novel and selective P2X3/P2X2/3 receptor antagonist, on neuropathic, inflammatory and chemogenic nociception following intrathecal and intraplantar administration. Br J Pharmacol 140:1381–8, 2003.CrossRefGoogle ScholarPubMed
Vergnolle, N, Bunnett, NW, Sharkey, KA, et al. Proteinase-activated receptor-2 and hyperalgesia: a novel pain pathway. Nat Med 7:821–6, 2001.CrossRefGoogle ScholarPubMed
Knotkova, H, Pappagallo, M. Pharmacology of pain transmission and modulation. II. Peripheral mechanisms. In: Pappagallo, M, ed. The neurological basis of pain. New York: McGraw-Hill, 2005, pp 53–60.Google Scholar
Ross, RA, Coutts, AA, McFarlane, SM et al. Actions of cannabinoid receptor ligands on rat cultured sensory neurones: implications for antinociception. Neuropharmacology 40:221–32, 2001.CrossRefGoogle ScholarPubMed
Piomelli, D, Beltramo, M, Giuffrida, A, et al. Endogenous cannabinoid signaling. Neurobiol Dis 5:462–73, 1998.CrossRefGoogle ScholarPubMed
Calignano, A, Rana, G, Giuffrida, A, et al. Control of pain initiation by endogenous cannabinoids. Nature 394:277–81, 1998.CrossRefGoogle ScholarPubMed
Davis, AJ, Perkins, MN. Induction of B1 receptors in vivo in a model of persistent inflammatory mechanical hyperalgesia in the rat. Neuropharmacology 33:127–33, 1994.CrossRefGoogle Scholar
Seabrook, GR, Bowery, BJ, Heavens, R, et al. Expression of B1 and B2 bradykinin receptor mRNA and their functional roles in sympathetic ganglia and sensory dorsal root ganglia neurones from wild-type and B2 receptor knockout mice. Neuropharmacology 36:1009–17, 1997.CrossRefGoogle ScholarPubMed
Gomes, JA, Li, X, Pan, HL et al. Intrathecal adenosine interacts with a spinal noradrenergic system to produce antinociception in nerve-injured rats. Anesthesiology 91:1072–9, 1999.CrossRefGoogle ScholarPubMed
Zhu, CZ, Mikusa, J, Chu, KL et al. A-134974: a novel adenosine kinase inhibitor, relieves tactile allodynia via spinal sites of action in a peripheral nerve injured rats. Brain Res 905:104–10, 2001.CrossRefGoogle Scholar
Sawynok, J. Adenosine receptor activation and nociception. Eur J Pharmacol 347:1–11, 1998.CrossRefGoogle ScholarPubMed
Pappagallo, M, Gaspardone, A, Tomai, F et al. Analgesic effect of bamiphylline on pain induced by intradermal injection of adenosine. Pain 53:199–204, 1993.CrossRefGoogle ScholarPubMed
Khandwala, H, Zhang, Z, Loomis, CW. Inhibition of strychnine-allodynia is mediated by spinal adenosine A1- but not A2-receptors in the rat. Brain Res 808:106–9, 1998.CrossRefGoogle Scholar
Matthews, EA, Dickenson, AH. Effects of spinally delivered N- and P-type voltage-dependent calcium channel antagonists on dorsal horn neuronal responses in a rat model of neuropathy. Pain 92:235–46, 2001.CrossRefGoogle Scholar
Luger, NM, Mach, DB, Sevcik, MA, et al. Bone cancer pain: from model to mechanism to therapy. J Pain Symptom Manage 29:S32–46, 2005.CrossRefGoogle Scholar
Lerner, UH. Neuropeptidergic regulation of bone resorption and bone formation. J Muskuloskelet Neuronal Interact 2:440–7, 2002.Google ScholarPubMed
Davis, JB, Gray, J, Gunthorpe, MJ, et al. Vanilloid receptor-1 is essential for inflammatory thermal hyperalgesia. Nature 405:183–7, 2000.CrossRefGoogle ScholarPubMed
Black, JA, Liu, S, Tanaka, M, et al. Changes in the expression of tetrodotoxin-sensitive sodium channels within dorsal root ganglia neurons in inflammatory pain. Pain 108:237–47, 2004.CrossRefGoogle ScholarPubMed
Waxman, SG, Dib-Hajj, SD. Erythromelalgia: a hereditary pain syndrome enters the molecular era. Ann Neurol 57:785–8, 2005.CrossRefGoogle ScholarPubMed
Marais, E, Klugbauer, N, Hofmann, F. Calcium channel alpha(2)delta subunits – structure and gabapentin binding. Mol Pharmacol 59:1243–8, 2001.CrossRefGoogle ScholarPubMed
Sutton, KG, Martin, DJ, Pinnock, RD. Gabapentin inhibits high-threshold calcium channel currents in cultured rat dorsal horn ganglion neurones. Br J Pharmacol 135:257–65, 2002.CrossRefGoogle Scholar
Lai, J, Hunter, JC, Porreca, F. The role of voltage-gated sodium channels in neuropathic pain. Curr Opin Neurobiol 13:291–7, 2003.CrossRefGoogle ScholarPubMed
Zhou, XF, Deng, YS, Xian, CJ, et al. Neurotrophins from dorsal root ganglia trigger allodynia after spinal nerve injury in rats. Eur J Neurosci 12:100–5, 2000.CrossRefGoogle ScholarPubMed
Schafers, M, Svensson, CI, Sommer, C, et al. Tumor necrosis factor-alpha induces mechanical allodynia after spinal nerve ligation by activation of p38 MAPK in primary sensory neurons. J Neurosci 23:2517–25, 2003.CrossRefGoogle ScholarPubMed
Jarvis, MF, Burgard, EC, McGaraughty, S, et al. A-317491, a novel potent and selective non-nucleotide antagonist of P2X3, and P2X2/3 receptors, reduces chronic inflammatory and neuropathic pain in the rat. Proc Natl Acad Sci U S A 99:17179–85, 2002.CrossRefGoogle ScholarPubMed
Boureau, F, Doubrere, JF, Luu, M. Study of verbal description in neuropathic pain. Pain 42:145–52, 1990.CrossRefGoogle ScholarPubMed
Farrar, JT, Portenoy, RK. Neuropathic cancer pain: the role of adjuvant analgesics. Oncology (Williston Park) 15:1435–42, 2001.Google ScholarPubMed
Deshpande, MA, Holden, RR, Gilron, I. The impact of therapy on quality of life and mood in neuropathic pain: what is the effect of pain reduction?Anesth Analg 102:1473–9, 2006.CrossRefGoogle ScholarPubMed
Ivez, R, Marsal, C, Vidal, J, et al. Cross-sectional evaluation of patient functioning and health-related quality of life in patients with neuropathic pain under standard care conditions. Eur J Pain 11:244–55, 2007.Google Scholar
Jensen, MP, Chodroff, MJ, Dworkin, RH. The impact of neuropathic pain on health-related quality of life: review and implications. Neurology 68:1178–82, 2007.CrossRefGoogle ScholarPubMed
Closs, SJ, Staples, V, Reid, I, et al. Managing the symptoms of neuropathic pain: an exploration of patients' experiences. J Pain Symptom Manage 34:422–33, 2007.CrossRefGoogle ScholarPubMed
Wallington, M, Mendis, S, Premawardhana, U, et al. Local control and survival in spinal cord compression from lymphoma and myeloma. Radiother Oncol 42:43–7, 1997.CrossRefGoogle ScholarPubMed
Correale, J, Monteverde, JA, Bueri, JA, et al. Peripheral nervous system and spinal cord involvement in lymphoma. Acta Neurol Scand 83:45–51, 1991.CrossRefGoogle ScholarPubMed
Vecht, CJ. Cancer pain: a neurological perspective. Curr Opin Neurol 13:649–53, 2000.CrossRefGoogle ScholarPubMed
Dimopoulos, MA, Alexanian, R. Waldenström's macroglobulinemia. Blood 83:1452–9, 1994.Google ScholarPubMed
Kissel, JT, Mendell, JR. Neuropathies associated with monoclonal gammopathies. Neuromuscul Disord 6:3–18, 1996.CrossRefGoogle ScholarPubMed
Wicklund, MP, Kissel, JT. Paraproteinemic neuropathy. Curr Treat Options Neurol 3:147–56, 2001.CrossRefGoogle ScholarPubMed
Portenoy, RK. Cancer pain – epidemiology and syndromes. Cancer 63:2298–307, 1989.3.0.CO;2-A>CrossRefGoogle ScholarPubMed
Chaudhry, V, Chaudhry, M, Crawford, TO, et al. Toxic neuropathy in patients with pre-existing neuropathy. Neurology 60:337–40, 2003.CrossRefGoogle ScholarPubMed
Hammack, JE, Michalak, JC, Loprinzi, CL, et al. Phase III evaluation of nortriptyline for alleviation of symptoms of cis-platinum-induced peripheral neuropathy. Pain 98:195–203, 2002.CrossRefGoogle ScholarPubMed
Postma, TJ, Hoekman, K, Riel, JM, et al. Peripheral neuropathy due to biweekly paclitaxel, epirubicin and cisplatin in patients with advanced ovarian cancer. J Neurooncol 45:241–6, 1999.CrossRefGoogle ScholarPubMed
Quasthoff, S, Hartung, HP. Chemotherapy-induced peripheral neuropathy. J Neurol 249:9–17, 2002.CrossRefGoogle ScholarPubMed
Grothey, A. Clinical management of oxaliplatin-associated neurotoxicity. Clin Colorectal Cancer 5(Suppl 1):S38–46, 2005.CrossRefGoogle ScholarPubMed
Lehky, TJ, Leonard, GD, Wilson, RH, et al. Oxaliplatin-induced neurotoxicity: acute hyperexcitability and chronic neuropathy. Muscle Nerve 29:387–92, 2004.CrossRefGoogle ScholarPubMed
Posner, JB. Side effects of chemotherapy. In: Posner, JB, ed. Neurologic complications of cancer. Philadelphia: F. A. Davis Company, 1995, pp 282–310.Google Scholar
Krishnan, AV, Goldstein, D, Friedlander, M, et al. Oxaliplatin-induced neurotoxicity and the development of neuropathy. Muscle Nerve 32:51–60, 2005.CrossRefGoogle ScholarPubMed
Verstappen, CC, Heimans, JJ, Hoekman, K, et al. Neurotoxic complications of chemotherapy in patients with cancer: clinical signs and optimal management. Drugs 63:1549–63, 2003.CrossRefGoogle ScholarPubMed
Argyriou, AA, Polychronopoulos, P, Koutras, A, et al. Peripheral neuropathy induced by administration of cisplatin- and paclitaxel-based chemotherapy. Could it be predicted?Support Care Cancer 13:647–651, 2005.CrossRefGoogle ScholarPubMed
Postma, TJ, Hoekman, K, Riel, JM, et al. Peripheral neuropathy due to biweekly paclitaxel, epirubicin and cisplatin in patients with advanced ovarian cancer. J Neurooncol 45:241–6, 1999.CrossRefGoogle ScholarPubMed
Chaudhry, V, Cornblath, DR, Corse, A, et al. Thalidomide-induced neuropathy. Neurology 59:1872–5, 2002.CrossRefGoogle ScholarPubMed
Visovsky, C. Chemotherapy-induced peripheral neuropathy. Cancer Invest 21:439–51, 2003.CrossRefGoogle ScholarPubMed
Gamelin, L, Boisdron-Celle, M, Delva, R, et al. Prevention of oxaliplatin-related neurotoxicity by calcium and magnesium infusions: a retrospective study of 161 patients receiving oxaliplatin combined with 5-fluorouracil and leucovorin for advanced colorectal cancer. Clin Cancer Res 10:4055–61, 2004.CrossRefGoogle ScholarPubMed
Cersosimo, RJ. Oxaliplatin-associated neuropathy: a review. Ann Pharmacother 39:128–35, 2005.CrossRefGoogle ScholarPubMed
Bove, L, Picardo, M, Maresca, V, et al. A pilot study on the relation between cisplatin neuropathy and vitamin E. J Exp Clin Cancer Res. 20:277–80, 2001.Google ScholarPubMed
Argyriou, AA, Chroni, E, Koutras, A, et al. Vitamin E for prophylaxis against chemotherapy-induced neuropathy: a randomized controlled trial. Neurology 64:26–31, 2005.CrossRefGoogle ScholarPubMed
Durand, JP, Alexandre, J, Guillevin, L, et al. Clinical activity of venlafaxine and topiramate against oxaliplatin-induced disabling permanent neuropathy. Anticancer Drugs 16:587–91, 2005.CrossRefGoogle ScholarPubMed
Birbaumer, N, Lutzenberg, W, Montoya, P, et al. Effects of regional anesthesia on phantom limb pain are mirrored in changes in cortical reorganization. J Neurosci 17:5503–8, 1997.CrossRefGoogle ScholarPubMed
Flor, H, Elbert, T, Knecht, S, et al. Phantom limb pain as a perceptual correlate of massive cortical reorganization following arm amputation. Nature 375:482–4, 1995.CrossRefGoogle Scholar
Karl, A, Birbaumer, N, Lutzenberger, W, et al. Reorganization of motor and somatosensory cortex in upper extremity amputees with phantom limb pain. J Neurosci 21:3609–18, 2001.CrossRefGoogle ScholarPubMed
Kew, JJ, Ridding, MC, Rothwell, JC, et al. Reorganization of cortical blood flow and transcranial magnetic stimulation maps in human subjects after upper limb amputation. J Neurophysiol 72:2517–24, 1994.CrossRefGoogle ScholarPubMed
Pascual-Leone, A, Peris, M, Tormos, JM, et al. Reorganization of human cortical motor output maps following traumatic forearm amputation. Neuroreport 7:2068–70, 1996.CrossRefGoogle ScholarPubMed
Sherman, RA, Arena, JG. Phantom limb pain: mechanisms, incidence and treatment. Clin Rev Phys Rehabil Med 4:1–26, 1992.Google Scholar
Jeannerod, M. The representing brain: neural correlates of motor intention and imagery. Brain Behav Sci 17:187–245, 1994.CrossRefGoogle Scholar
Jeannerod, M. Mental imagery in the motor context. Neuropsychologia 33:1419–32, 1995.CrossRefGoogle ScholarPubMed
Lotze, M, Grodd, W, Birbaumer, N, et al. Does the use of a myoelectric prosthesis prevent cortical reorganization and phantom limb pain?Nat Neurosci 2:501–2, 1999.CrossRefGoogle ScholarPubMed
Lotze, M, Flor, H, Grodd, W, et al. Phantom movements and pain. An fMRI study in upper limb amputees. Brain 124:2268–77, 2001.CrossRefGoogle ScholarPubMed
Pezzin, , Dillingham, TR, MacKenzie, EJ. Rehabilitation and the long-term outcomes of persons with trauma-related amputations. Arch Phys Med Rehabil 81:292–300, 2000.CrossRefGoogle ScholarPubMed
Johnson, R. Herpes zoster – predicting and minimizing the impact of post-herpetic neuralgia. J Antimicrob Chemother Suppl T1:1–8, 2001.Google Scholar
Haanpaa, M, Laippala, P, Nurmikko, T. Allodynia and pinprick hypesthesia in acute herpes zoster, and the development of postherpetic neuralgia. J Pain Symptom Manage. 20:50–8, 2000.CrossRefGoogle ScholarPubMed
Tremonts-Lukats, IW, Megeff, C, Backonja, MM. Anticonvulsants for neuropathic pain syndromes: mechanisms of action and place in therapy. Drugs 60:1029–52, 2000.CrossRefGoogle Scholar
Backonja, M, Beydoun, A, Edwards, KR, et al. Gabapentin for the symptomatic treatment of painful neuropathy in patients with diabetes mellitus: a randomized controlled trial. JAMA 280:1831–6, 1998.CrossRefGoogle ScholarPubMed
Rowbotham, M, Harden, N, Stacey, B, et al. Gabapentin for the treatment of postherpetic neuralgia: a randomized controlled trial. JAMA 280:1837–42, 1998.CrossRefGoogle ScholarPubMed
Caraceni, A, Zecca, E, Martini, C, et al. Gabapentin as an adjuvant to opioid analgesia for neuropathic cancer pain. J Pain Symptom Manage 17:441–5, 1999.CrossRefGoogle ScholarPubMed
Bosnjak, S, Jelic, S, Susnjar, S, et al. Gabapentin for relief of neuropathic pain related to anticancer treatment: a preliminary study. J Chemother 14:214–19, 2002.CrossRefGoogle ScholarPubMed
Twycross, R, Wilcock, A: Palliative care formulary, v2.0 CD-ROM. 2002. Palliativedrugs.com.
Gilron, I, Bailey, JM, Tu, D, et al. Morphine, gabapentin, or their combination for neuropathic pain. N Engl J Med 352:1324–34, 2005.CrossRefGoogle ScholarPubMed
Richter, RW, Portenoy, R, Sharma, U, et al. Relief of painful diabetic peripheral neuropathy with pregabalin: a randomized, placebo-controlled trial. J Pain 6:253–60, 2005.CrossRefGoogle ScholarPubMed
Freynhagen, R, Strojek, K, Griesing, T, et al. Efficacy of pregabalin in neuropathic pain evaluated in a 12-week, randomised, double-blind, multicentre, placebo-controlled trial of flexible- and fixed-dose regimens. Pain 115:254–63, 2005.CrossRefGoogle Scholar
Hadj Tahar, A. Pregabalin for peripheral neuropathic pain. Issues Emerg Health Technol March:1–4, 2005.Google ScholarPubMed
Freynhagen, R, Grond, S, Hagebeuker, A, et al. Efficacy and safety of pregabalin in treatment of refractory patients with various neuropathic pain entities in clinical routine. Int J Clin Pract 61:1989–96, 2007.CrossRefGoogle ScholarPubMed
Brodie, MJ. Lamotrigine. Lancet 339:1397–400, 1992.CrossRefGoogle ScholarPubMed
Guay, DR. Oxcarbazepine, topiramate, zonisamide, and levetiracetam: potential use in neuropathic pain. Am J Geriatr Pharmacother 1:18–37, 2003.CrossRefGoogle ScholarPubMed
Nakamura-Craig, M, Follenfant, RL. Effect of lamotrigine in the effect of acute and chronic hyperalgesia induced by PGE2 and in the chronic hyperalgesia in rats with streptozotocin-induced diabetes. Pain 63:33–7, 1995.CrossRefGoogle ScholarPubMed
Canavero, S, Bonicalzi, V. Lamotrigine control of central pain. Pain 68:179–81, 1996.CrossRefGoogle ScholarPubMed
Vestergaard, K, Andersen, G, Gottrup, H, et al. Lamotrigine for central poststroke pain: a randomized controlled trial. Neurology 56:184–90, 2001.CrossRefGoogle ScholarPubMed
Mockenhaupt, M, Messenheimer, J, Tennis, P, et al. Risk of Stevens-Johnson syndrome and toxic epidermal necrolysis in new users of antiepileptics. Neurology 64:1134–8, 2005.CrossRefGoogle ScholarPubMed
Vinik, AI, Tuchman, M, Safirstein, B, et al. Lamotrigine for treatment of pain associated with diabetic neuropathy: results of two randomized, double-blind, placebo-controlled studies. Pain 128:168–79, 2007.CrossRefGoogle ScholarPubMed
Silver, M, Blum, D, Grainger, J, et al. Double blind, placebo-controlled trial of lamotrigine in combination with other medication for neuropathic pain. J Pain Symptom Manage 34:446–54, 2007.CrossRefGoogle ScholarPubMed
Breuer, B, Pappagallo, M, Knotkova, H, et al. A randomized, double-blind, placebo-controlled, two-period, crossover, pilot trial of lamotrigine in patients with central pain due to multiple sclerosis. Clin Ther 29:2022–30, 2007.CrossRefGoogle ScholarPubMed
Sobotka, JL, Alexander, B, Cook, BL. A review of carbamazepine's hematologic reactions and monitoring recommendations. DICP 24:1214–19, 1990.CrossRefGoogle ScholarPubMed
Portenoy, RK, Khan, E, Layman, M, et al. Chronic morphine therapy for cancer pain: plasma and cerebrospinal fluid morphine and morphine-6-glucuronide concentrations. Neurology 41:1457–61, 1991.CrossRefGoogle ScholarPubMed
Jadad, AR. The WHO anelgesic ladder for cancer pain management: stepping up the quality of its evaluation. JAMA 274, 1870–73, 1995.CrossRefGoogle Scholar
Trujillo, KA, Akil, H. Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801. Science 251:85–7, 1991.CrossRefGoogle ScholarPubMed
Dole, VP. Implications of methadone maintenance for theories of narcotic addiction. JAMA 260:3025–9, 1988.CrossRefGoogle ScholarPubMed
Nyswander, M, Dole, VP. The present status of methadone blockade treatment. Am J Psychiatry 123:1441–2, 1967.CrossRefGoogle ScholarPubMed
Krantz, MJ, Kutinsky, IB, Robertson, AD, et al. Dose-related effects of methadone on QT prolongation in a series of patients with torsade de pointes. Pharmacotherapy 23:802–5, 2003.CrossRefGoogle Scholar
Cruciani, RA, Sekine, R, Homel, P, et al. Measurements of QTc in patients receiving chronic methadone therapy. J Pain Symptom Manage 29:385–91, 2005.CrossRefGoogle Scholar
Kornick, CA, Kilborn, MJ, Santiago-Palma, J, et al. QTc interval prolongation associated with intravenous methadone. Pain 105:499–506, 2003.CrossRefGoogle ScholarPubMed
Mercadante, S. Opioid rotation for cancer pain: rationale and clinical aspects. Cancer 86:1856–66, 1999.3.0.CO;2-G>CrossRefGoogle ScholarPubMed
Mercadante, S, Cacuccio, A, Fulfaro, F, et al. Switching from morphine to methadone to improve analgesia and tolerability in cancer patients: a prospective study. J Clin Oncol 19:2898–904, 2001.CrossRefGoogle ScholarPubMed
Max, MB, Lynch, SA, Muir, J, et al. Effects of desipramine, amitriptyline, and fluoxetine on pain in diabetic neuropathy. N Engl J Med 326:1250–6, 1992.CrossRefGoogle ScholarPubMed
Watson, CP, Vernich, L, Chipman, M, et al. Nortriptyline versus amitriptyline in postherpetic neuralgia: a randomized trial. Neurology 51:1166, 1998.CrossRefGoogle ScholarPubMed
Morello, CM, Lechband, SG, Stoner, CP, et al. Randomized double blind study comparing the efficacy of gabapentin with amitriptyline in diabetic peripheral neuropathy pain. Arch Intern Med 59:1931–7, 1999.CrossRefGoogle Scholar
Kalso, E, Tasmuth, T, Neuvonen, P. Amitriptyline effectively relieves neuropathic pain following treatment of breast cancer. Pain 64:293–302, 1996.Google ScholarPubMed
Ansari, A. The efficacy of newer antidepressants in the treatment of chronic pain: a review of current literature. Harv Rev Psychiatry 7:257–77, 2000.CrossRefGoogle ScholarPubMed
Sindrup, SH, Grodum, E, Gram, LF, et al. Concentration-response relationship in paroxetine treatment of diabetic neuropathy symptoms: a patient-blinded dose-escalation study. Ther Drug Monit 13:408–14, 1991.CrossRefGoogle ScholarPubMed
Semenchuk, MR, Sherman, S, Davis, B. Double-blind, randomized trial of bupropion SR for the treatment of neuropathic pain. Neurology 57:1583–8, 2001.CrossRefGoogle ScholarPubMed
Goldstein, DJ, Lu, Y, Detke, MJ, et al. Duloxetine vs. placebo in patients with painful diabetic neuropathy. Pain 116:109–18, 2005.CrossRefGoogle ScholarPubMed
Raskin, J, Pritchett, YL, Wang, F, et al. A double blind, randomized multicenter trial comparing duloxetine with placebo in the management of diabetic peripheral neuropathic pain. Pain Med 6:346–56, 2005.CrossRefGoogle ScholarPubMed
Wernicke, JF, Pritchett, YL, D'Souza, DN, et al. A randomized controlled trial of duloxetine in diabetic peripheral neuropathic pain. Neurology 67:1411–20, 2006.CrossRefGoogle ScholarPubMed
Rowbotham, MC, Goli, V, Kunz, NR, Lei, D. Venlafaxine extended release in the treatment of painful diabetic neuropathy: a double-blind, placebo controlled study. Pain 110:697–706, 2004.CrossRefGoogle ScholarPubMed
Mao, J, Chen, LL. Systemic lidocaine for neuropathic pain relief. Pain 87:7–17, 2000.CrossRefGoogle ScholarPubMed
Galer, BS, Harle, J, Rowbotham, MC. Response to intravenous lidocaine infusion predicts subsequent response to oral mexiletine: a prospective study. J Pain Symptom Manage 12:161–7, 1996.CrossRefGoogle ScholarPubMed
Chong, SF, Bretscher, ME, Mailliard, JA, et al. Pilot study evaluating local anesthetics administered systemically for treatment of pain in patients with advanced cancer. J Pain Symptom Manage 13:112–17, 1997.CrossRefGoogle ScholarPubMed
Bennett, G, Serafini, M, Burchiel, K, et al. Evidence-based review of the literature on intrathecal delivery of pain medication. J Pain Symptom Manage 20:S12–36, 2000.CrossRefGoogle ScholarPubMed
Du Pen, SL, Kharash, ED, Williams, A, et al. Chronic epidural bupivacaine-opioid infusion in intractable cancer pain. Pain 49:293–300, 1992.CrossRefGoogle ScholarPubMed
Slonimski, M, Abram, SE, Zuniga, RE. Intrathecal baclofen in pain management. Reg Anesth Pain Med 29:269–76, 2004.CrossRefGoogle ScholarPubMed
Kotani, N, Kushikata, T, Hashimoto, H, et al. Intrathecal methylprednisolone for intractable postherpetic neuralgia. N Engl J Med 343:1514–19, 2000.CrossRefGoogle ScholarPubMed
Pierce, TL, Tiong, GK, Olley, JE. Morphine and methadone dependence in the rat: withdrawal and brain metenkephalin levels. Pharmacol Biochem Behav 42:91–6, 1992.CrossRefGoogle Scholar
Kannan, TR, Saxena, A, Bhatnagar, S, et al. Oral ketamine as an adjuvant to oral morphine for neuropathic pain in cancer patients. J Pain Symptom Manage 23:60–5, 2002.CrossRefGoogle ScholarPubMed
Kiefer, RT, Rohr, P, Ploppa, A, et al. Efficacy of ketamine in anesthetic dosage for the treatment of refractory complex regional pain syndrome: an open label phase II study. Pain Med 9:1173–201, 2008.CrossRefGoogle Scholar
Koffler, SP, Hampstead, BM, Irani, F, et al. The neurocognitive effects of 5 day anesthetic ketamine for the treatment of refractory complex regional pain syndrome. Arch Clin Neuropsychol 22:719–29, 2007.CrossRefGoogle ScholarPubMed
Cvrcek, P. Side effects of ketamine in the long-term treatment of neuropathic pain. Pain Med 9:253–7, 2008.CrossRefGoogle ScholarPubMed
Fromm, GH. Baclofen as an adjuvant analgesic. J Pain Symptom Manage 9:500–9, 1994.CrossRefGoogle ScholarPubMed
Fernandez, F, Adams, F, Holmes, VF. Analgesic effect of alprazolam in patients with chronic, organic pain of malignant origin. J Clin Psychopharmacol 7:167–9, 1987.CrossRefGoogle ScholarPubMed
Pavlakis, N, Schmidt, R, Stockler, M. Bisphosphonates for breast cancer. Cochrane Database Syst Rev CD003474, 2005.CrossRefGoogle Scholar
Terpos, E, Dimopoulos, MA. Myeloma bone disease: pathophysiology and management. Ann Oncol 16:1223–31, 2005.CrossRefGoogle ScholarPubMed
Reszka, AA, Rodan, GA. Mechanism of action of bisphosphonates. Curr Osteoporos Rep 1:45–52, 2003.CrossRefGoogle ScholarPubMed
Kovcin, V, Jelić, S, Babović, N, Tomasević, Z. A pilot study to assess the efficacy of salmon calcitonin in the relief of neuropathic pain caused by extraskeletal metastases. Support Care Cancer 2:71–3, 1994.CrossRefGoogle ScholarPubMed
Vecht, CJ, Haaxma-Reiche, H, van Putten, WL, et al. Initial bolus of conventional versus high-dose dexamethasone in metastatic spinal cord compression. Neurology 39:1255–7, 1989.CrossRefGoogle ScholarPubMed
Watanabe, S, Bruera, E. Corticosteroids as adjuvant analgesics. J Pain Symptom Manage 9:442–5, 1994.CrossRefGoogle ScholarPubMed
Khojainova, N, Santiago-Palma, J, Kornick, C, et al. Olanzapine in the management of cancer pain. J Pain Symptom Manage 23:346–50, 2002.CrossRefGoogle ScholarPubMed
Hohmann, AG. Spinal and peripheral mechanisms of cannabinoid antinociception: behavioral, neurophysiological and neuroanatomical perspectives. Chem Phys Lipids 121:173–90, 2002.CrossRefGoogle ScholarPubMed
Goya, P, Jagerovic, N, Hernandez-Folgado, L, et al. Cannabinoids and neuropathic pain. Mini Rev Med Chem 3:765–72, 2003.CrossRefGoogle ScholarPubMed
Cravatt, BF, Lichtman, AH. The endogenous cannabinoid system and its role in nociceptive behavior. J Neurobiol 61:149–60, 2004.CrossRefGoogle ScholarPubMed
Hoffman, AG. Spinal and peripheral mechanisms of cannabinoid antinociception: behavioral, neurophysiological and neuroanatomical perspectives. Chem Phys Lipids 121:173–90, 2002.Google Scholar
Karst, M, Salim, K, Burstein, S. Analgesic affect of the synthetic cannabinoid CT-3 on chronic neuropathic pain: a randomized controlled trial. JAMA 290:1757–62, 2003.CrossRefGoogle Scholar
Nurmikko, TJ, Serpell, MG, Hoggart, B, et al. Sativex successfully treats neuropathic pain characterized by allodynia: a randomized, double-blind, placebo-controlled clinical trial. Pain 133:210–20, 2007.CrossRefGoogle Scholar
Galer, BS, Rowbotham, MC, Perander, J, et al. Topical lidocaine patch relieves postherpetic neuralgia more effectively than a vehicle topical patch: results of an enriched enrollment study. Pain 80:533–8, 1999.CrossRefGoogle ScholarPubMed
White, WT, Patel, N, Drass, M, et al. Lidocaine patch 5% with systemic analgesics such as gabapentin: a rational polypharmacy approach for the treatment of chronic pain. Pain Med. (4):321–30, 2003.CrossRefGoogle ScholarPubMed
Ellison, N, Loprinzi, CL, Kugler, J, et al. Phase III placebo-controlled trial of capsaicin cream in the management of surgical neuropathic pain in cancer patients. J Clin Oncol 15:2974–80, 1997.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×