10,435
Views
48
CrossRef citations to date
0
Altmetric
Review

Perispinal etanercept: a new therapeutic paradigm in neurology

Pages 985-1002 | Published online: 09 Jan 2014

References

  • Clark IA. How TNF was recognized as a key mechanism of disease. Cytokine Growth Factor Rev.18(3–4), 335–343 (2007).
  • Tracey D, Klareskog L, Sasso EH, Salfeld JG, Tak PP. Tumor necrosis factor antagonist mechanisms of action: a comprehensive review. Pharmacol. Ther.117(2), 244–279 (2008).
  • Tarkowski E, Blennow K, Wallin A, Tarkowski A. Intracerebral production of tumor necrosis factor-α, a local neuroprotective agent, in Alzheimer disease and vascular dementia. J. Clin. Immunol.19(4), 223–230 (1999).
  • Tarkowski E, Andreasen N, Tarkowski A, Blennow K. Intrathecal inflammation precedes development of Alzheimer’s disease. J. Neurol. Neurosurg. Psychiatry74(9), 1200–1205 (2003).
  • Tobinick E. Tumour necrosis factor modulation for treatment of Alzheimer’s disease: rationale and current evidence. CNS Drugs23(9), 713–725 (2009).
  • Buchhave P, Zetterberg H, Blennow K, Minthon L, Janciauskiene S, Hansson O. Soluble TNF receptors are associated with Ab metabolism and conversion to dementia in subjects with mild cognitive impairment. Neurobiol. Aging DOI: 10.1016/j.neurobiolaging.2008.10.012 (2008) (Epub ahead of print).
  • Ferguson AR, Christensen RN, Gensel JC et al. Cell death after spinal cord injury is exacerbated by rapid TNF α-induced trafficking of GluR2-lacking AMPARs to the plasma membrane. J. Neurosci.28(44), 11391–11400 (2008).
  • McCoy MK, Tansey MG. TNF signaling inhibition in the CNS: implications for normal brain function and neurodegenerative disease. J. Neuroinflammation5, 45 (2008).
  • Sjogren M, Folkesson S, Blennow K, Tarkowski E. Increased intrathecal inflammatory activity in frontotemporal dementia: pathophysiological implications. J. Neurol. Neurosurg. Psychiatry75(8), 1107–1111 (2004).
  • Tweedie D, Sambamurti K, Greig NH. TNF-α inhibition as a treatment strategy for neurodegenerative disorders: new drug candidates and targets. Curr. Alzheimer Res.4(4), 378–385 (2007).
  • Uceyler N, Sommer C. Cytokine regulation in animal models of neuropathic pain and in human diseases. Neurosci. Lett.437(3), 194–198 (2008).
  • Van Eldik LJ, Thompson WL, Ranaivo HR, Behanna HA, Watterson DM. Proinflammatory cytokine upregulation as a therapeutic target for neurodegenerative diseases: function-based and target-based discovery approaches. Int. Rev. Neurobiol.82, 278–297 (2007).
  • Halassa MM, Fellin T, Haydon PG. The tripartite synapse: roles for gliotransmission in health and disease. Trends Mol. Med.13(2), 54–63 (2007).
  • Bains JS, Oliet SH. Glia: they make your memories stick! Trends Neurosci.30(8), 417–424 (2007).
  • Oliet SH, Piet R, Poulain DA, Theodosis DT. Glial modulation of synaptic transmission: insights from the supraoptic nucleus of the hypothalamus. Glia47(3), 258–267 (2004).
  • De Lella Ezcurra AL, Chertoff M, Ferrari C, Graciarena M, Pitossi F. Chronic expression of low levels of tumor necrosis factor-α in the substantia nigra elicits progressive neurodegeneration, delayed motor symptoms and microglia/macrophage activation. Neurobiol. Dis.37(3), 630–640 (2010).
  • Floden AM, Li S, Combs CK. β-amyloid-stimulated microglia induce neuron death via synergistic stimulation of tumor necrosis factor a and NMDA receptors. J. Neurosci.25(10), 2566–2575 (2005).
  • Gorlovoy P, Larionov S, Pham TT, Neumann H. Accumulation of tau induced in neurites by microglial proinflammatory mediators. FASEB J.23(8), 2502–2513 (2009).
  • Janelsins MC, Mastrangelo MA, Oddo S, LaFerla FM, Federoff HJ, Bowers WJ. Early correlation of microglial activation with enhanced tumor necrosis factor-α and monocyte chemoattractant protein-1 expression specifically within the entorhinal cortex of triple transgenic Alzheimer’s disease mice. J. Neuroinflammation2, 23 (2005).
  • Mrak RE, Griffin WS. Glia and their cytokines in progression of neurodegeneration. Neurobiol. Aging26(3), 349–354 (2005).
  • Ranaivo HR, Craft JM, Hu W et al. Glia as a therapeutic target: selective suppression of human amyloid-β-induced upregulation of brain proinflammatory cytokine production attenuates neurodegeneration. J. Neurosci.26(2), 662–670 (2006).
  • Van Eldik LJ, Thompson WL, Ralay Ranaivo H, Behanna HA, Martin Watterson D. Glia proinflammatory cytokine upregulation as a therapeutic target for neurodegenerative diseases: function-based and target-based discovery approaches. Int. Rev. Neurobiol.82, 277–296 (2007).
  • Tobinick E. Perispinal etanercept for neuroinflammatory disorders. Drug Discov. Today14(3–4), 168–177 (2009).
  • Tobinick EL, Britschgi-Davoodifar S. Perispinal TNF-α inhibition for discogenic pain. Swiss Med. Wkly133(11–12), 170–177 (2003).
  • Sommer C, Schafers M. Mechanisms of neuropathic pain: the role of cytokines. Drug Discov. Today1(4), 441–448 (2004).
  • Myers RR, Campana WM, Shubayev VI. The role of neuroinflammation in neuropathic pain: mechanisms and therapeutic targets. Drug Discov. Today11(1–2), 8–20 (2006).
  • Homma Y, Brull SJ, Zhang JM. A comparison of chronic pain behavior following local application of tumor necrosis factor a to the normal and mechanically compressed lumbar ganglia in the rat. Pain95(3), 239–246 (2002).
  • Onda A, Hamba M, Yabuki S, Kikuchi S. Exogenous tumor necrosis factor-α induces abnormal discharges in rat dorsal horn neurons. Spine27(15), 1618–1624; discussion 1624 (2002).
  • Aoki Y, Rydevik B, Kikuchi S, Olmarker K. Local application of disc-related cytokines on spinal nerve roots. Spine (Phila Pa 1976)27(15), 1614–1617 (2002).
  • Sorkin LS, Doom CM. Epineurial application of TNF elicits an acute mechanical hyperalgesia in the awake rat. J. Peripher. Nerv. Syst.5(2), 96–100 (2000).
  • Igarashi T, Kikuchi S, Shubayev V, Myers RR. 2000 Volvo Award winner in basic science studies: exogenous tumor necrosis factor-α mimics nucleus pulposus-induced neuropathology. Molecular, histologic, and behavioral comparisons in rats. Spine (Phila Pa 1976)25(23), 2975–2980 (2000).
  • Olmarker K, Rydevik B. Selective inhibition of tumor necrosis factor-α prevents nucleus pulposus-induced thrombus formation, intraneural edema, and reduction of nerve conduction velocity: possible implications for future pharmacologic treatment strategies of sciatica. Spine (Phila Pa 1976)26(8), 863–869 (2001).
  • Murata Y, Olmarker K, Takahashi I, Takahashi K, Rydevik B. Effects of selective tumor necrosis factor-α inhibition to pain-behavioral changes caused by nucleus pulposus-induced damage to the spinal nerve in rats. Neurosci. Lett.382(1–2), 148–152 (2005).
  • Olmarker K, Nutu M, Storkson R. Changes in spontaneous behavior in rats exposed to experimental disc herniation are blocked by selective TNF-α inhibition. Spine28(15), 1635–1641; discussion 1642 (2003).
  • Sekiguchi M, Kikuchi S, Myers RR. Experimental spinal stenosis: relationship between degree of cauda equina compression, neuropathology, and pain. Spine (Phila Pa 1976)29(10), 1105–1111 (2004).
  • Igarashi A, Kikuchi S, Konno S, Olmarker K. Inflammatory cytokines released from the facet joint tissue in degenerative lumbar spinal disorders. Spine29(19), 2091–2095 (2004).
  • Sommer C, Schafers M, Marziniak M, Toyka KV. Etanercept reduces hyperalgesia in experimental painful neuropathy. J. Peripher. Nerv. Syst.6(2), 67–72 (2001).
  • Kato K, Kikuchi S, Shubayev VI, Myers RR. Distribution and tumor necrosis factor-α isoform binding specificity of locally administered etanercept into injured and uninjured rat sciatic nerve. Neuroscience160(2), 492–500 (2009).
  • Kato K, Liu H, Kikuchi SI, Myers RR, Shubayev VI. Immediate anti-tumor necrosis factor-α (etanercept) therapy enhances axonal regeneration after sciatic nerve crush. J. Neurosci. Res.88(2), 360–368 (2009).
  • Tobinick E, Vega CP. The cerebrospinal venous system: anatomy, physiology, and clinical implications. MedGenMed8(1), 53 (2006).
  • Batson OV. The function of the vertebral veins and their role in the spread of metastases. Ann. Surg.112, 138–149 (1940).
  • Batson OV. The vertebral vein system. Caldwell lecture, 1956. Am. J. Roentgenol. Radium Ther. Nucl. Med.78(2), 195–212 (1957).
  • Tobinick EL. Targeted etanercept for discogenic neck pain: uncontrolled, open-label results in two adults. Clin. Ther.25(4), 1211–1218 (2003).
  • Tobinick E, Davoodifar S. Efficacy of etanercept delivered by perispinal administration for chronic back and/or neck disc-related pain: a study of clinical observations in 143 patients. Curr. Med. Res. Opin.20(7), 1075–1085 (2004).
  • Kume K, Amano K, Yamada S. The efficacy and safety of caudal epidural injection with the TNF-α antagonist, etanercept, in patients with disc-herniation-induced sciatica: results of a randomized, controlled, 1-month follow-up study. Ann. Rheum. Dis.67(Suppl. II), 131 (2008).
  • Shin K, Lee S, Moon S et al. A prospective controlled trial of TNF-α inhibitor for symptomatic patients with cervical disk herniation. Spine J.5(4), S45 (2005).
  • Genevay S, Stingelin S, Gabay C. Efficacy of etanercept in the treatment of acute, severe sciatica: a pilot study. Ann. Rheum. Dis.63(9), 1120–1123 (2004).
  • Cohen SP, Bogduk N, Dragovich A et al. Randomized, double-blind, placebo-controlled, dose-response, and preclinical safety study of transforaminal epidural etanercept for the treatment of sciatica. Anesthesiology110(5), 1116–1126 (2009).
  • Kume K, Amano A, Yamada S, Nagata H. The efficacy and safety of caudal epidural injection with the TNF-αntagonist, adalimumab and etanercept, in patients with disc-herniation-induced sciatica. Results of a randomized, controlled, 1-month follow-up study. Presented at: 2009 Annual Meeting of the American College of Rheumatology. Philadelphia, PA, USA, 17–21 October 2009.
  • Furst DE, Keystone EC, Fleischmann R et al. Updated consensus statement on biological agents for the treatment of rheumatic diseases, 2009. Ann. Rheum. Dis.69(Suppl. 1), i2–i29 (2010).
  • Payne R, Inturrisi CE. CSF distribution of morphine, methadone and sucrose after intrathecal injection. Life Sci.37(12), 1137–1144 (1985).
  • Rafii MS, Aisen PS. Recent developments in Alzheimer’s disease therapeutics. BMC Med.7, 7 (2009).
  • Querfurth HW, LaFerla FM. Alzheimer’s Disease. N. Engl. J. Med.362(4), 329–344 (2010).
  • Alkam T, Nitta A, Mizoguchi H et al. Restraining tumor necrosis factor-α by thalidomide prevents the amyloid β-induced impairment of recognition memory in mice. Behav. Brain Res.189(1), 100–106 (2008).
  • Giuliani F, Vernay A, Leuba G, Schenk F. Decreased behavioral impairments in an Alzheimer mice model by interfering with TNF-α metabolism. Brain Res. Bull.80(4–5), 302–308 (2009).
  • Hu NW, Klyubin I, Anwy R, Rowan MJ. GluN2B subunit-containing NMDA receptor antagonists prevent Aβ-mediated synaptic plasticity disruption in vivo. Proc. Natl Acad. Sci. USA106(48), 20504–20509 (2009).
  • Medeiros R, Figueiredo CP, Pandolfo P et al. The role of TNF-α signaling pathway on COX-2 upregulation and cognitive decline induced by β-amyloid peptide. Behav. Brain Res.209(1), 165–173 (2010).
  • Medeiros R, Prediger RD, Passos GF et al. Connecting TNF-α signaling pathways to iNOS expression in a mouse model of Alzheimer’s disease: relevance for the behavioral and synaptic deficits induced by amyloid b protein. J. Neurosci.27(20), 5394–5404 (2007).
  • Rowan MJ, Klyubin I, Wang Q, Hu NW, Anwyl R. Synaptic memory mechanisms: Alzheimer’s disease amyloid β-peptide-induced dysfunction. Biochem. Soc. Trans.35(Pt 5), 1219–1223 (2007).
  • Wang Q, Wu J, Rowan MJ, Anwyl R. β-amyloid inhibition of long-term potentiation is mediated via tumor necrosis factor. Eur J. Neurosci.22(11), 2827–2832 (2005).
  • Tobinick E. Perispinal etanercept for treatment of Alzheimer’s disease. Curr. Alzheimer Res.4(5), 550–552 (2007).
  • Tobinick EL, Gross H. Rapid cognitive improvement in Alzheimer’s disease following perispinal etanercept administration. J. Neuroinflammation5, 2 (2008).
  • Yamamoto M, Kiyota T, Horiba M et al. Interferon-γ and tumor necrosis factor-α regulate amyloid-β plaque deposition and β-secretase expression in Swedish mutant APP transgenic mice. Am. J. Pathol.170(2), 680–692 (2007).
  • Liao YF, Wang BJ, Cheng HT, Kuo LH, Wolfe MS. Tumor necrosis factor-α, interleukin-1β, and Interferon-γ stimulate γ-secretase-mediated cleavage of amyloid precursor protein through a JNK-dependent MAPK pathway. J. Biol. Chem.279(47), 49523–49532 (2004).
  • Combs CK, Karlo JC, Kao SC, Landreth GE. β-amyloid stimulation of microglia and monocytes results in TNFα-dependent expression of inducible nitric oxide synthase and neuronal apoptosis. J. Neurosci.21(4), 1179–1188 (2001).
  • Ishii W, Kishida D, Suzuki A et al. A case with rheumatoid arthritis and systemic reactive AA amyloidosis showing rapid regression of amyloid deposition on gastroduodenal mucosa after a combined therapy of corticosteroid and etanercept. Rheumatol. Int. DOI: 10.007.600296-009-1205-2 (2009) (Epub ahead of print).
  • Hussein MA, Juturi JV, Rybicki L, Lutton S, Murphy BR, Karam MA. Etanercept therapy in patients with advanced primary amyloidosis. Med. Oncol.20(3), 283–290 (2003).
  • Salloway S, Sperling R, Gilman S et al. A Phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology73(24), 2061–2070 (2009).
  • Cacci E, Ajmone-Cat MA, Anelli T, Biagioni S, Minghetti L. In vitro neuronal and glial differentiation from embryonic or adult neural precursor cells are differently affected by chronic or acute activation of microglia. Glia56(4), 412–425 (2008).
  • Holmes C, Cunningham C, Zotova E et al. Systemic inflammation and disease progression in Alzheimer disease. Neurology73(10), 768–774 (2009).
  • Akiyama H, Barger S, Barnum S et al. Inflammation and Alzheimer’s disease. Neurobiol. Aging21(3), 383–421 (2000).
  • Mori T, Koyama N, Arendash GW, Horikoshi-Sakuraba Y, Tan J, Town T. Overexpression of human S100B exacerbates cerebral amyloidosis and gliosis in the Tg2576 mouse model of Alzheimer’s disease. Glia58(3), 300–314 (2010).
  • Perry RT, Collins JS, Wiener H, Acton R, Go RC. The role of TNF and its receptors in Alzheimer’s disease. Neurobiol. Aging22(6), 873–883 (2001).
  • Alvarez V, Mata IF, Gonzalez P et al. Association between the TNFα-308 A/G polymorphism and the onset-age of Alzheimer disease. Am. J. Med. Genet.114(5), 574–577 (2002).
  • Tan ZS, Beiser AS, Vasan RS et al. Inflammatory markers and the risk of Alzheimer disease: the Framingham Study. Neurology68(22), 1902–1908 (2007).
  • Tarkowski E, Liljeroth AM, Minthon L, Tarkowski A, Wallin A, Blennow K. Cerebral pattern of pro- and anti-inflammatory cytokines in dementias. Brain Res. Bull.61(3), 255–260 (2003).
  • van Exel E, Eikelenboom P, Comijs H et al. Vascular factors and markers of inflammation in offspring with a parental history of late-onset Alzheimer disease. Arch. Gen. Psychiatry66(11), 1263–1270 (2009).
  • Park KM, Bowers WJ. Tumor necrosis factor-α mediated signaling in neuronal homeostasis and dysfunction. Cell. Signal.22(7), 977–983 (2010).
  • Di Bona D, Candore G, Franceschi C et al. Systematic review by meta-analyses on the possible role of TNF-α polymorphisms in association with Alzheimer’s disease. Brain Res. Rev.61(2), 60–68 (2009).
  • Laws SM, Perneczky R, Wagenpfeil S et al. TNF polymorphisms in Alzheimer disease and functional implications on CSF β-amyloid levels. Hum. Mutat.26(1), 29–35 (2005).
  • Lio D, Annoni G, Licastro F et al. Tumor necrosis factor-α -308A/G polymorphism is associated with age at onset of Alzheimer’s disease. Mech. Ageing Dev.127(6), 567–571 (2006).
  • Ramos EM, Lin MT, Larson EB et al. Tumor necrosis factor a and interleukin 10 promoter region polymorphisms and risk of late-onset Alzheimer disease. Arch. Neurol.63(8), 1165–1169 (2006).
  • Zuliani G, Ranzini M, Guerra G et al. Plasma cytokines profile in older subjects with late onset Alzheimer’s disease or vascular dementia. J. Psychiatr. Res.41(8), 686–693 (2006).
  • Tobinick E, Gross H, Weinberger A, Cohen H. TNF-α modulation for treatment of Alzheimer’s disease: a 6-month pilot study. MedGenMed8(2), 25 (2006).
  • Tobinick EL, Gross H. Rapid improvement in verbal fluency and aphasia following perispinal etanercept in Alzheimer’s disease. BMC Neurol.8, 27 (2008).
  • Tobinick E. Perispinal etanercept produces rapid improvement in primary progressive aphasia: identification of a novel, rapidly reversible TNF-mediated pathophysiologic mechanism. Medscape J. Med.10(6), 135 (2008).
  • Groen RJ, du Toit DF, Phillips FM et al. Anatomical and pathological considerations in percutaneous vertebroplasty and kyphoplasty: a reappraisal of the vertebral venous system. Spine (Phila Pa 1976)29(13), 1465–1471 (2004).
  • Vallejo MC, Beaman ST, Ramanathan S. Blurred vision as the only symptom of a positive epidural test dose. Anesth. Analg.102(3), 973–974 (2006).
  • Anderson R. Diodrast studies of the vertebral and cranial venous systems to show their probable role in cerebral metastases. J. Neurosurg.8(4), 411–422 (1951).
  • Hassler O. Deep cerebral venous system in man. A microangiographic study on its areas of drainage and its anastomoses with the superficial cerebral veins. Neurology16(5), 505–511 (1966).
  • Wacnik PW, Eikmeier LJ, Simone DA, Wilcox GL, Beitz AJ. Nociceptive characteristics of tumor necrosis factor-α in naive and tumor-bearing mice. Neuroscience132(2), 479–491 (2005).
  • Lorberboym M, Lampl Y, Sadeh M. Correlation of 99mTc-DTPA SPECT of the blood–brain barrier with neurologic outcome after acute stroke. J. Nucl. Med.44(12), 1898–1904 (2003).
  • Bohac D, Burke W, Cotter R, Zheng J, Potter J, Gendelman H. A 24-week randomized, double-blind, placebo-controlled study of the efficacy and tolerability of TNFR: Fc (etanercept) in the treatment of dementia of the Alzheimer type. Neurobiol. Aging23(1 Suppl. 1), S1–S606 (2002) (Abstract 315).
  • Cao Q, Cai W, Li ZB et al. PET imaging of acute and chronic inflammation in living mice. Eur. J. Nucl. Med. Mol. Imaging34(11), 1832–1842 (2007).
  • Tobinick EL, Chen K, Chen X. Rapid intracerebroventricular delivery of Cu-DOTA-etanercept after peripheral administration demonstrated by PET imaging. BMC Res. Notes2, 28 (2009).
  • Maness LM, Banks WA, Zadina JE, Kastin AJ. Selective transport of blood-borne interleukin-1 a into the posterior division of the septum of the mouse brain. Brain Res.700(1–2), 83–88 (1995).
  • Pfefferbaum A, Adalsteinsson E, Sullivan EV. In vivo structural imaging of the rat brain with a 3-T clinical human scanner. J. Magn. Reson. Imaging20(5), 779–785 (2004).
  • Quan N, Mhlanga JD, Whiteside MB, McCoy AN, Kristensson K, Herkenham M. Chronic overexpression of proinflammatory cytokines and histopathology in the brains of rats infected with Trypanosoma brucei. J. Comp. Neurol.414(1), 114–130 (1999).
  • Yanamoto K, Zhang MR, Kumata K, Hatori A, Okada M, Suzuki K. In vitro and ex vivo autoradiography studies on peripheral-type benzodiazepine receptor binding using [11C]AC-5216 in normal and kainic acid-lesioned rats. Neurosci. Lett.428(2–3), 59–63 (2007).
  • Johanson CE, Duncan JA, Stopa EG, Baird A. Enhanced prospects for drug delivery and brain targeting by the choroid plexus–CSF route. Pharm. Res.22(7), 1011–1037 (2005).
  • Starr JM, Farrall AJ, Armitage P, McGurn B, Wardlaw J. Blood–brain barrier permeability in Alzheimer’s disease: a case–control MRI study. Psychiatry Res.171(3), 232–241 (2009).
  • Poduslo JF, Curran GL, Wengenack TM, Malester B, Duff K. Permeability of proteins at the blood–brain barrier in the normal adult mouse and double transgenic mouse model of Alzheimer’s disease. Neurobiol. Dis.8(4), 555–567 (2001).
  • Clark SR, McMahon CJ, Gueorguieva I et al. Interleukin-1 receptor antagonist penetrates human brain at experimentally therapeutic concentrations. J. Cereb. Blood Flow Metab.28(2), 387–394 (2008).
  • Ransohoff RM, Kivisakk P, Kidd G. Three or more routes for leukocyte migration into the central nervous system. Nat. Rev. Immunol.3(7), 569–581 (2003).
  • Johanson CE, Duncan JA 3rd, Klinge PM, Brinker T, Stopa EG, Silverberg GD. Multiplicity of cerebrospinal fluid functions: new challenges in health and disease. Cerebrospinal Fluid Res.5, 10 (2008).
  • Serot JM, Bene MC, Faure GC. Choroid plexus, aging of the brain, and Alzheimer’s disease. Front. Biosci.8, S515–S521 (2003).
  • Griffin WS. Perispinal etanercept: potential as an Alzheimer therapeutic. J. Neuroinflammation5, 3 (2008).
  • Pelletier M, Siegel RM. Wishing away inflammation? New links between serotonin and TNF signaling. Mol. Interv.9(6), 299–301 (2009).
  • Chavant F, Deguil J, Pain S et al. Imipramine, in part through tumor necrosis factor a inhibition, prevents cognitive decline and β-amyloid accumulation in a mouse model of Alzheimer’s disease. J. Pharmacol. Exp. Ther.332(2), 505–514 (2010).
  • Wheeler D, Knapp E, Bandaru VV et al. Tumor necrosis factor-α-induced neutral sphingomyelinase-2 modulates synaptic plasticity by controlling the membrane insertion of NMDA receptors. J. Neurochem.109(5), 1237–1249 (2009).
  • Nygard M, Lundkvist GB, Hill RH, Kristensson K. Rapid nitric oxide-dependent effects of tumor necrosis factor-α on suprachiasmatic nuclei neuronal activity. Neuroreport20(2), 213–217 (2009).
  • Wang Y. P4-266: modification of synaptic plasticity by TNF and sphingomyelinase: implications for cognitive impairment in Alzheimer’s disease. Alzheimers Dement.4(4 Suppl.), T749 (2008).
  • Wolinsky H. Paths to acceptance. The advancement of scientific knowledge is an uphill struggle against ‘accepted wisdom’. EMBO Rep.9(5), 416–418 (2008).
  • Youn DH, Wang H, Jeong SJ. Exogenous tumor necrosis factor-α rapidly alters synaptic and sensory transmission in the adult rat spinal cord dorsal horn. J. Neurosci. Res.86(13), 2867–2875 (2008).
  • Maarouf CL, Daugs ID, Spina S et al. Histopathological and molecular heterogeneity among individuals with dementia associated with Presenilin mutations. Mol. Neurodegener.3, 20 (2008).
  • Passant U, Rosen I, Gustafson L, Englund E. The heterogeneity of frontotemporal dementia with regard to initial symptoms, qEEG and neuropathology. Int. J. Geriatr. Psychiatry20(10), 983–988 (2005).
  • Mayeux R, Stern Y, Spanton S. Heterogeneity in dementia of the Alzheimer type: evidence of subgroups. Neurology35(4), 453–461 (1985).
  • Schneider JA, Arvanitakis Z, Bang W, Bennett DA. Mixed brain pathologies account for most dementia cases in community-dwelling older persons. Neurology69(24), 2197–2204 (2007).
  • Cedazo-Minguez A, Winblad B. Biomarkers for Alzheimer’s disease and other forms of dementia: clinical needs, limitations and future aspects. Exp. Gerontol.45(1), 5–14 (2009).
  • Fagan AM, Csernansky CA, Morris JC, Holtzman DM. The search for antecedent biomarkers of Alzheimer’s disease. J. Alzheimers Dis.8(4), 347–358 (2005).
  • Halperin I, Morelli M, Korczyn AD, Youdim MB, Mandel SA. Biomarkers for evaluation of clinical efficacy of multipotential neuroprotective drugs for Alzheimer’s and Parkinson’s diseases. Neurotherapeutics6(1), 128–140 (2009).
  • Wilcock GK. Bapineuzumab in Alzheimer’s disease: where now? Lancet Neurol.9(2), 134–136 (2010).
  • Beattie EC, Stellwagen D, Morishita W et al. Control of synaptic strength by glial TNFα. Science295(5563), 2282–2285 (2002).
  • Stellwagen D, Malenka RC. Synaptic scaling mediated by glial TNF-α. Nature440(7087), 1054–1059 (2006).
  • Stellwagen D, Beattie EC, Seo JY, Malenka RC. Differential regulation of AMPA receptor and GABA receptor trafficking by tumor necrosis factor-α. J. Neurosci.25(12), 3219–3228 (2005).
  • Nasreddine ZS, Phillips NA, Bedirian V et al. The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive impairment. J. Am. Geriatr. Soc.53(4), 695–699 (2005).
  • Vellas B. Tarenflurbil for Alzheimer’s disease: a “shot on goal” that missed. Lancet Neurol.9(3), 235–237 (2010).
  • Harrison JE, Buxton P, Husain M, Wise R. Short test of semantic and phonological fluency: normal performance, validity and test–retest reliability. Br. J. Clin. Psychol.39(Pt 2), 181–191 (2000).
  • Fisher NJ, Tierney MC, Rourke BP, Szalai JP. Verbal fluency patterns in two subgroups of patients with Alzheimer’s disease. Clin. Neuropsychol.18(1), 122–131 (2004).
  • Simonsick EM, Gardner AW, Poehlman ET. Assessment of physical function and exercise tolerance in older adults: reproducibility and comparability of five measures. Aging (Milano)12(4), 274–280 (2000).
  • Tyring S, Gottlieb A, Papp K et al. Etanercept and clinical outcomes, fatigue, and depression in psoriasis: double-blind placebo-controlled randomised Phase III trial. Lancet367(9504), 29–35 (2006).
  • Apkarian AV, Sosa Y, Sonty S et al. Chronic back pain is associated with decreased prefrontal and thalamic gray matter density. J. Neurosci.24(46), 10410–10415 (2004).
  • Dowlati Y, Herrmann N, Swardfager W et al. A meta-analysis of cytokines in major depression. Biol. Psychiatry67(5), 446–457 (2009).
  • Amgen Inc., Thousand Oaks, California, data on file (2010).
  • Beck JM, Azari ED. FDA, off-label use, and informed consent: debunking myths and misconceptions. Food Drug Law J.53(1), 71–104 (1998).
  • Bermpohl D, You Z, Lo EH, Kim HH, Whalen MJ. TNF a and Fas mediate tissue damage and functional outcome after traumatic brain injury in mice. J. Cereb. Blood Flow Metab.27(11), 1806–1818 (2007).
  • Liesz A, Suri-Payer E, Veltkamp C et al. Regulatory T cells are key cerebroprotective immunomodulators in acute experimental stroke. Nat. Med.15(2), 192–199 (2009).
  • Marousi S, Ellul J, Karakantza M. Genetic polymorphisms of type-1 and type-2 inflammatory cytokines in ischaemic stroke. Vasc. Dis. Prev.5(2), 89–103 (2008).
  • Zaremba J, Losy J. Early TNF-α levels correlate with ischaemic stroke severity. Acta Neurol. Scand.104(5), 288–295 (2001).
  • Zaremba J, Skrobanski P, Losy J. Tumour necrosis factor-α is increased in the cerebrospinal fluid and serum of ischaemic stroke patients and correlates with the volume of evolving brain infarct. Biomed. Pharmacother.55(5), 258–263 (2001).
  • Hayashi M, Ueyama T, Nemoto K, Tamaki T, Senba E. Sequential mRNA expression for immediate early genes, cytokines, and neurotrophins in spinal cord injury. J. Neurotrauma17(3), 203–218 (2000).
  • Hermann GE, Rogers RC, Bresnahan JC, Beattie MS. Tumor necrosis factor-α induces cFOS and strongly potentiates glutamate-mediated cell death in the rat spinal cord. Neurobiol. Dis.8(4), 590–599 (2001).
  • Yune TY, Chang MJ, Kim SJ et al. Increased production of tumor necrosis factor-α induces apoptosis after traumatic spinal cord injury in rats. J. Neurotrauma20(2), 207–219 (2003).
  • Kurt G, Ergun E, Cemil B et al. Neuroprotective effects of infliximab in experimental spinal cord injury. Surg. Neurol.71(3), 332–336; discussion 336 (2009).
  • Peng XM, Zhou ZG, Glorioso JC, Fink DJ, Mata M. Tumor necrosis factor-α contributes to below-level neuropathic pain after spinal cord injury. Ann. Neurol.59(5), 843–851 (2006).
  • Sharma HS, Winkler T, Stalberg E, Gordh T, Alm P, Westman J. Topical application of TNF-α antiserum attenuates spinal cord trauma induced edema formation, microvascular permeability disturbances and cell injury in the rat. Acta Neurochir. Suppl.86, 407–413 (2003).
  • Genovese T, Mazzon E, Crisafulli C et al. Immunomodulatory effects of etanercept in an experimental model of spinal cord injury. J. Pharmacol. Exp. Ther.316(3), 1006–1016 (2006).
  • Genovese T, Mazzon E, Crisafulli C et al. Combination of dexamethasone and etanercept reduces secondary damage in experimental spinal cord trauma. Neuroscience150(1), 168–181 (2007).
  • Marchand F, Tsantoulas C, Singh D et al. Effects of etanercept and minocycline in a rat model of spinal cord injury. Eur. J. Pain13(7), 673–681 (2009).
  • Dinomais M, Stana L, Egon G, Richard I, Menei P. Significant recovery of motor function in a patient with complete T7 paraplegia receiving etanercept. J. Rehabil. Med.41(4), 286–288 (2009).
  • Chi LY, Yu J, Zhu H, Li XG, Zhu SG, Kindy MS. The dual role of tumor necrosis factor-α in the pathophysiology of spinal cord injury. Neurosci. Lett.438(2), 174–179 (2008).
  • Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat. Rev. Neurosci.9(1), 46–56 (2008).
  • Pardridge WM. The blood–brain barrier: bottleneck in brain drug development. NeuroRx2(1), 3–14 (2005).
  • Banks WA. Are the extracellular pathways a conduit for the delivery of therapeutics to the brain? Curr. Pharm. Des.10(12), 1365–1370 (2004).
  • Banks WA, Plotkin SR, Kastin AJ. Permeability of the blood–brain barrier to soluble cytokine receptors. Neuroimmunomodulation2(3), 161–165 (1995).
  • Rubenstein JL, Combs D, Rosenberg J et al. Rituximab therapy for CNS lymphomas: targeting the leptomeningeal compartment. Blood101(2), 466–468 (2003).
  • Rubenstein JL, Fridlyand J, Abrey L et al. Phase I study of intraventricular administration of rituximab in patients with recurrent CNS and intraocular lymphoma. J. Clin. Oncol.25(11), 1350–1356 (2007).
  • Boettger MK, Weber K, Grossmann D et al. Spinal TNF-α neutralization reduces peripheral inflammation and hyperalgesia and suppresses autonomic responses in experimental arthritis. Arthritis Rheum. DOI: 10.1002/art.27380 (2010) (Epub ahead of print).
  • Tobinick E. Spinal delivery of p38: TNF-α inhibitors. PLoS Med.3(11), e511 (2006).
  • Tobinick EL. Targeted etanercept for treatment-refractory pain due to bone metastasis: two case reports. Clin. Ther.25(8), 2279–2288 (2003).
  • Tobinick EL. A critique of intradiscal administration for treatment of radiculopathy. Anesthesiology108(2), 334; author reply 335 (2008).
  • Tancredi V, D’Arcangelo G, Grassi F et al. Tumor necrosis factor alters synaptic transmission in rat hippocampal slices. Neurosci. Lett.146(2), 176–178 (1992).
  • Netter F. The CIBA Collection of Medical Illustrations. Volume 1: Nervous System; Part I: Anatomy and Physiology. Ciba Pharmaceutical Products, Inc., NJ, USA (1953).
  • Dahl E, Cohen SP. Perineural injection of etanercept as a treatment for postamputation pain. Clin. J. Pain24(2), 172–175 (2008).
  • Cohen SP, Wenzell D, Hurley RW et al. A double-blind, placebo-controlled, dose–response pilot study evaluating intradiscal etanercept in patients with chronic discogenic low back pain or lumbosacral radiculopathy. Anesthesiology107(1), 99–105 (2007).
  • Malik K. Epidural etanercept for lumbar radiculopathy. Anaesth. Intensive Care35(2), 301–302 (2007).
  • Serratrice J, de Roux-Serratrice C, Disdier P, Dode C, Weiller PJ. Dramatic etanercept-induced remission of relapsing febrile sciatic neuralgia related to p46l mutation of the tnfrsf1a gene. Clin. Rheumatol.26(9), 1535–1536 (2007).
  • McAlpine FE, Tansey MG. Neuroinflammation and tumor necrosis factor signaling in the pathophysiology of Alzheimer’s. J. Inflam. Res.1, 29–39 (2008).
  • McNaull BB, Todd S, McGuinness B, Passmore AP. Inflammation and anti-inflammatory strategies for Alzheimer’s disease – a mini-review. Gerontology56(1), 3–14 (2009).
  • Teeling JL, Perry VH. Systemic infection and inflammation in acute CNS injury and chronic neurodegeneration: underlying mechanisms. Neuroscience158(3), 1062–1073 (2009)

Website

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.