SCISSOR-Spinal Cord Injury Study on Small molecule-derived Rho inhibition: a clinical study protocol

Marcel A Kopp, Thomas Liebscher, Ralf Watzlawick, Peter Martus, Stefan Laufer, Christian Blex, Ralf Schindler, Gerhard J Jungehulsing, Sven Knüppel, Martin Kreutzträger, Axel Ekkernkamp, Ulrich Dirnagl, Stephen M Strittmatter, Andreas Niedeggen, Jan M Schwab, Marcel A Kopp, Thomas Liebscher, Ralf Watzlawick, Peter Martus, Stefan Laufer, Christian Blex, Ralf Schindler, Gerhard J Jungehulsing, Sven Knüppel, Martin Kreutzträger, Axel Ekkernkamp, Ulrich Dirnagl, Stephen M Strittmatter, Andreas Niedeggen, Jan M Schwab

Abstract

Introduction: The approved analgesic and anti-inflammatory drugs ibuprofen and indometacin block the small GTPase RhoA, a key enzyme that impedes axonal sprouting after axonal damage. Inhibition of the Rho pathway in a central nervous system-effective manner requires higher dosages compared with orthodox cyclooxygenase-blocking effects. Preclinical studies on spinal cord injury (SCI) imply improved motor recovery after ibuprofen/indometacin-mediated Rho inhibition. This has been reassessed by a meta-analysis of the underlying experimental evidence, which indicates an overall effect size of 20.2% regarding motor outcome achieved after ibuprofen/indometacin treatment compared with vehicle controls. In addition, ibuprofen/indometacin may also limit sickness behaviour, non-neurogenic systemic inflammatory response syndrome (SIRS), neuropathic pain and heterotopic ossifications after SCI. Consequently, 'small molecule'-mediated Rho inhibition after acute SCI warrants clinical investigation.

Methods and analysis: Protocol of an investigator-initiated clinical open-label pilot trial on high-dose ibuprofen treatment after acute traumatic, motor-complete SCI. A sample of n=12 patients will be enrolled in two cohorts treated with 2400 mg/day ibuprofen for 4 or 12 weeks, respectively. The primary safety end point is an occurrence of serious adverse events, primarily gastroduodenal bleedings. Secondary end points are pharmacokinetics, feasibility and preliminary effects on neurological recovery, neuropathic pain and heterotopic ossifications. The primary safety analysis is based on the incidence of severe gastrointestinal bleedings. Additional analyses will be mainly descriptive and casuistic.

Ethics and dissemination: The clinical trial protocol was approved by the responsible German state Ethics Board, and the Federal Institute for Drugs and Medical Devices. The study complies with the Declaration of Helsinki, the principles of Good Clinical Practice and all further applicable regulations. This safety and pharmacokinetics trial informs the planning of a subsequent randomised controlled trial. Regardless of the result of the primary and secondary outcome assessments, the clinical trial will be reported as a publication in a peer-reviewed journal.

Trial registration number: NCT02096913; Pre-results.

Keywords: Heterotopic ossifications; Neuropathic pain; Neuroprotection; Plasticity; ibuprofen.

Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/

Figures

Figure 1
Figure 1
Pharmacological targets of ibuprofen. Intracellular signalling cascades converge at the GTPase RhoA, which is activated after SCI by myelin and scar-associated proteins (for review, see refs. 5, 7 and 29). Downstream to Rho, the activated ROCK inhibits axonal regrowth, promotes neurodegeneration, contributes to the development of neuropathic pain and tissue loss and impedes neurorestoration and functional recovery (reviewed by Watzlawick et al). This pathway can be blocked by the ROCK inhibitors Y-27632 and fasudil or the specific Rho inhibitors P21CIP1/WAF1, C3 transferase, and by the R(−) and S(−) enantiomers of ibuprofen, as the most convincing Rho inhibitor among individual drugs from the group of NSAIDs. Ibuprofen-mediated Rho inhibition depends on the upregulation of PPARγ. Treatment with PPARγ agonists was demonstrated to have anti-inflammatory effects and to protect tissue and thereby motor function in other CNS injury conditions (reviewed by McTigue). It is not yet clear whether the inhibition of NF-κB as a further target of R(−)/S(+) ibuprofen is independent of PPARγ. Notably, PPARγ inhibits gene expression by antagonising the activities of the proinflammatory transcription factors NF-κB. Another pathway, mainly operated by the S enantiomer of ibuprofen, is the inhibition of COX 1/2 and consequently of the prostaglandin E2 production, which activates NF-κB or counterregulates it at very high concentrations. COX 1/2 and NF-κB are associated with inflammation-induced neuropathic pain, neurodegeneration, sickness behaviour and the systemic inflammatory response syndrome. Systemic inflammation contributes to neurogenic heterotopic ossificastions. Taken together, Rho-blocking NSAIDs have the potential to decrease the systemic and acute CNS inflammatory response by targeting at least two separate pathways, PPARγ and COX 1/2. The suspected side effect of neuroprotective anti-inflammatory therapy, that is, that it further limits the regeneration capacity of spared axons, is suggested to be abrogated by Rho inhibition. CNS, central nervous system; COX, cyclooxygenase; GDP, guanosine diphosphate; GTP, guanosine triphosphate; NSAIDs, non-steroid anti-inflammatory drugs; PPARγ, peroxisome proliferator-activated receptor γ; NF-κB, nuclear factor κB; RGMa, repulsive guidance molecule A; ROCK, Rho-associated coiled kinase; SCI, spinal cord injury.
Figure 2
Figure 2
Systematic review preclinical study selection chart. To identify animal studies reporting the effect of ibuprofen or indometacin treatment for neurobehavioral recovery after SCI, the following search term was used for PubMed, EMBASE and ISI Web of science (search conducted on 18 May 2015): (Ibuprofen OR Indometacin OR NSAID OR nonsteroidal anti-inflammatory drugs) AND (SCI OR hemisection OR contusion OR dorsal column injury OR transection OR corticospinal tract injury OR compression OR spinal cord lesion). Search results were limited to animals. Studies were included if they reported the effects of ibuprofen or indometacin in animal models after various types of SCIs. We included SCI experiments comparing functional motor outcome between a group of animals receiving treatment and a control group receiving no treatment (sham group). Non-traumatic models of SCI were excluded, as well as studies reporting only combined treatments. Studies had to report the number of animals for each group, the mean effect size and its variance. Studies were excluded due to inappropriate outcome scales, combination of treatments and statistical inconsistencies. SCI, spinal cord injury.
Figure 3
Figure 3
Meta-analysis of preclinical effects on motor recovery. Improvement in neurobehavioral score is ranked by effect size (ES). The overall number of included animals was n=255 (median n=12, range 8–73). Black dots represent studies using ibuprofen and white dots show indometacin studies. The horizontal bar represents the 95% CI of the ES. Details on the design of the included studies are summarised in table 1.
Figure 4
Figure 4
Longitudinal clinical trial design. Diagram of frequency and scope of trial procedures. The evaluation for eligibility should start as early as possible after acute SCI. The baseline will be obtained at the day of the inclusion from day 4 and latest at day 21 post trauma, in any case as early as possible. Start of the study medication is directly after the baseline assessment. The duration of the intervention is 4 weeks for cohort I and 12 weeks for cohort II. Frequent safety laboratory measurements are performed. Samples for pharmacokinetic measurements are collected two times in cohort I and three times in cohort II. The follow-up visits for the determination of secondary end points are performed at week 4 (±3 days) and after the end of the intervention at week 24 (±14 days). Final safety laboratory measurements will be performed 4 weeks after the end of the study medication. SCI, spinal cord injury.

References

    1. Hurlbert RJ, Hadley MN, Walters BC et al. . Pharmacological therapy for acute spinal cord injury. Neurosurgery 2013;72(Suppl 2):93–105. 10.1227/NEU.0b013e31827765c6
    1. Siddiqui AM, Khazaei M, Fehlings MG. Translating mechanisms of neuroprotection, regeneration, and repair to treatment of spinal cord injury. Prog Brain Res 2015;218:15–54. 10.1016/bs.pbr.2014.12.007
    1. Dietz V, Curt A. Translating preclinical approaches into human application. Handb Clin Neurol 2012;109:399–409. 10.1016/B978-0-444-52137-8.00025-5
    1. Thuret S, Moon LD, Gage FH. Therapeutic interventions after spinal cord injury. Nat Rev Neurosci 2006;7:628–43. 10.1038/nrn1955
    1. Yiu G, He Z. Glial inhibition of CNS axon regeneration. Nat Rev Neurosci 2006;7:617–27. 10.1038/nrn1956
    1. Xie F, Zheng B. White matter inhibitors in CNS axon regeneration failure. Exp Neurol 2008;209:302–12. 10.1016/j.expneurol.2007.07.005
    1. Kopp MA, Liebscher T, Niedeggen A et al. . Small-molecule-induced Rho-inhibition: NSAIDs after spinal cord injury. Cell Tissue Res 2012;349:119–32. 10.1007/s00441-012-1334-7
    1. Schmandke A, Schmandke A, Strittmatter SM. ROCK and Rho: biochemistry and neuronal functions of Rho-associated protein kinases. Neuroscientist 2007;13:454–69. 10.1177/1073858407303611
    1. Baer AS, Syed YA, Kang SU et al. . Myelin-mediated inhibition of oligodendrocyte precursor differentiation can be overcome by pharmacological modulation of Fyn-RhoA and protein kinase C signalling. Brain 2009;132(Pt 2):465–81. 10.1093/brain/awn334
    1. Tanaka H, Yamashita T, Yachi K et al. . Cytoplasmic p21(Cip1/WAF1) enhances axonal regeneration and functional recovery after spinal cord injury in rats. Neuroscience 2004;127:155–64. 10.1016/j.neuroscience.2004.05.010
    1. Wang X, Budel S, Baughman K et al. . Ibuprofen enhances recovery from spinal cord injury by limiting tissue loss and stimulating axonal growth. J Neurotrauma 2009;26:81–95. 10.1089/neu.2007.0464
    1. Wu D, Yang P, Zhang X et al. . Targeting a dominant negative rho kinase to neurons promotes axonal outgrowth and partial functional recovery after rat rubrospinal tract lesion. Mol Ther 2009;17: 2020–30. 10.1038/mt.2009.168
    1. Boato F, Hendrix S, Huelsenbeck SC et al. . C3 peptide enhances recovery from spinal cord injury by improved regenerative growth of descending fiber tracts. J Cell Sci 2010;123(Pt 10):1652–62. 10.1242/jcs.066050
    1. Boomkamp SD, Riehle MO, Wood J et al. . The development of a rat in vitro model of spinal cord injury demonstrating the additive effects of Rho and ROCK inhibitors on neurite outgrowth and myelination. Glia 2012;60:441–56. 10.1002/glia.22278
    1. Chan CC, Khodarahmi K, Liu J et al. . Dose-dependent beneficial and detrimental effects of ROCK inhibitor Y27632 on axonal sprouting and functional recovery after rat spinal cord injury. Exp Neurol 2005;196:352–64. 10.1016/j.expneurol.2005.08.011
    1. Dergham P, Ellezam B, Essagian C et al. . Rho signaling pathway targeted to promote spinal cord repair. J Neurosci 2002;22:6570–7.
    1. Dill J, Patel AR, Yang XL et al. . A molecular mechanism for ibuprofen-mediated RhoA inhibition in neurons. J Neurosci 2010;30:963–72. 10.1523/JNEUROSCI.5045-09.2010
    1. Fournier AE, Takizawa BT, Strittmatter SM. Rho kinase inhibition enhances axonal regeneration in the injured CNS. J Neurosci 2003;23:1416–23.
    1. Fu Q, Hue J, Li S. Nonsteroidal anti-inflammatory drugs promote axon regeneration via RhoA inhibition. J Neurosci 2007;27:4154–64. 10.1523/JNEUROSCI.4353-06.2007
    1. Otsuka S, Adamson C, Sankar V et al. . Delayed intrathecal delivery of RhoA siRNA to the contused spinal cord inhibits allodynia, preserves white matter, and increases serotonergic fiber growth. J Neurotrauma 2011;28:1063–76. 10.1089/neu.2010.1568
    1. Roloff F, Scheiblich H, Dewitz C et al. . Enhanced neurite outgrowth of human model (NT2) neurons by small-molecule inhibitors of Rho/ROCK signaling. PLoS ONE 2015;10(2):e0118536 10.1371/journal.pone.0118536
    1. Loske P, Boato F, Hendrix S et al. . Minimal essential length of Clostridium botulinum C3 peptides to enhance neuronal regenerative growth and connectivity in a non-enzymatic mode. J Neurochem 2012;120:1084–96. 10.1111/j.1471-4159.2012.07657.x
    1. Nishio Y, Koda M, Kitajo K et al. . Delayed treatment with Rho-kinase inhibitor does not enhance axonal regeneration or functional recovery after spinal cord injury in rats. Exp Neurol 2006;200:392–7. 10.1016/j.expneurol.2006.02.123
    1. Lord-Fontaine S, Yang F, Diep Q et al. . Local inhibition of Rho signaling by cell-permeable recombinant protein BA-210 prevents secondary damage and promotes functional recovery following acute spinal cord injury. J Neurotrauma 2008;25:1309–22. 10.1089/neu.2008.0613
    1. Xing B, Li H, Wang H et al. . RhoA-inhibiting NSAIDs promote axonal myelination after spinal cord injury. Exp Neurol 2011;231:247–60. 10.1016/j.expneurol.2011.06.018
    1. Schwab JM, Hirsch S, Monnier PP et al. . The Rho-GTPase inhibitor C3-C2IN/C2II induces functional neuronal recovery in a rat model of severe spinal cord injury. Society for Neuroscience, 2002.
    1. Mueller BK, Mack H, Teusch N. Rho kinase, a promising drug target for neurological disorders. Nat Rev Drug Discov 2005;4:387–98. 10.1038/nrd1719
    1. Watzlawick R, Sena ES, Dirnagl U et al. . Effect and reporting bias of RhoA/ROCK-blockade intervention on locomotor recovery after spinal cord injury: a systematic review and meta-analysis. JAMA Neurol 2014;71:91–9. 10.1001/jamaneurol.2013.4684
    1. McKerracher L, Higuchi H. Targeting Rho to stimulate repair after spinal cord injury. J Neurotrauma 2006;23:309–17. 10.1089/neu.2006.23.309
    1. McKerracher L, Anderson KD. Analysis of recruitment and outcomes in the phase I/IIa Cethrin clinical trial for acute spinal cord injury. J Neurotrauma 2013;30:1795–804. 10.1089/neu.2013.2909
    1. Zhou Y, Su Y, Li B et al. . Nonsteroidal anti-inflammatory drugs can lower amyloidogenic Abeta42 by inhibiting Rho. Science 2003;302:1215–17. 10.1126/science.1090154
    1. Sharp KG, Yee KM, Stiles TL et al. . A re-assessment of the effects of treatment with a non-steroidal anti-inflammatory (ibuprofen) on promoting axon regeneration via RhoA inhibition after spinal cord injury. Exp Neurol 2013;248:321–37. 10.1016/j.expneurol.2013.06.023
    1. Ramirez SH, Heilman D, Morsey B et al. . Activation of peroxisome proliferator-activated receptor gamma (PPARgamma) suppresses Rho GTPases in human brain microvascular endothelial cells and inhibits adhesion and transendothelial migration of HIV-1 infected monocytes. J Immunol 2008;180:1854–65. 10.4049/jimmunol.180.3.1854
    1. Jaradat MS, Wongsud B, Phornchirasilp S et al. . Activation of peroxisome proliferator-activated receptor isoforms and inhibition of prostaglandin H(2) synthases by ibuprofen, naproxen, and indomethacin. Biochem Pharmacol 2001;62:1587–95. 10.1016/S0006-2952(01)00822-X
    1. Cragg JJ, Haefeli J, Jutzeler CR et al. . Effects of pain and pain management on motor recovery of spinal cord-injured patients: a longitudinal study. Neurorehabil Neural Repair 2016. doi: 10.1177/1545968315624777 [epub ahead of print 7 Jan 2016]10.1177/1545968315624777
    1. Lehmann JM, Lenhard JM, Oliver BB et al. . Peroxisome proliferator-activated receptors alpha and gamma are activated by indomethacin and other non-steroidal anti-inflammatory drugs. J Biol Chem 1997;272:3406–10. 10.1074/jbc.272.6.3406
    1. Crofford LJ. COX-1 and COX-2 tissue expression: implications and predictions. J Rheumatol Suppl 1997;49:15–19.
    1. Scheuren N, Bang H, Munster T et al. . Modulation of transcription factor NF-kappaB by enantiomers of the nonsteroidal drug ibuprofen. Br J Pharmacol 1998;123:645–52. 10.1038/sj.bjp.0701652
    1. Ricote M, Li AC, Willson TM et al. . The peroxisome proliferator-activated receptor-gamma is a negative regulator of macrophage activation. Nature 1998;391:79–82. 10.1038/34178
    1. Jiang C, Ting AT, Seed B. PPAR-gamma agonists inhibit production of monocyte inflammatory cytokines. Nature 1998;391:82–6. 10.1038/34184
    1. McTigue DM. Potential therapeutic targets for PPARgamma after spinal cord injury. PPAR Res 2008;2008:517162 10.1155/2008/517162
    1. Schwab JM, Brechtel K, Nguyen TD et al. . Persistent accumulation of cyclooxygenase-1 (COX-1) expressing microglia/macrophages and upregulation by endothelium following spinal cord injury. J Neuroimmunol 2000;111:122–30. 10.1016/S0165-5728(00)00372-6
    1. Teeling JL, Cunningham C, Newman TA et al. . The effect of non-steroidal anti-inflammatory agents on behavioural changes and cytokine production following systemic inflammation: implications for a role of COX-1. Brain Behav Immun 2010;24:409–19. 10.1016/j.bbi.2009.11.006
    1. O'Brien AJ, Fullerton JN, Massey KA et al. . Immunosuppression in acutely decompensated cirrhosis is mediated by prostaglandin E2. Nat Med 2014;20:518–23. 10.1038/nm.3516
    1. Stables MJ, Newson J, Ayoub SS et al. . Priming innate immune responses to infection by cyclooxygenase inhibition kills antibiotic-susceptible and -resistant bacteria. Blood 2010;116:2950–9. 10.1182/blood-2010-05-284844
    1. Meisel C, Schwab JM, Prass K et al. . Central nervous system injury-induced immune deficiency syndrome. Nat Rev Neurosci 2005;6:775–86. 10.1038/nrn1765
    1. Schwab JM, Zhang Y, Kopp MA et al. . The paradox of chronic neuroinflammation, systemic immune suppression, autoimmunity after traumatic chronic spinal cord injury. Exp Neurol 2014;258:121–9. 10.1016/j.expneurol.2014.04.023
    1. Failli V, Kopp MA, Gericke C et al. . Functional neurological recovery after spinal cord injury is impaired in patients with infections. Brain 2012;135(Pt 11):3238–50. 10.1093/brain/aws267
    1. Bethea JR, Castro M, Keane RW et al. . Traumatic spinal cord injury induces nuclear factor-kappaB activation. J Neurosci 1998;18:3251–60.
    1. Frakes AE, Ferraiuolo L, Haidet-Phillips AM et al. . Microglia induce motor neuron death via the classical NF-kappaB pathway in amyotrophic lateral sclerosis. Neuron 2014;81:1009–23. 10.1016/j.neuron.2014.01.013
    1. Moreno B, Jukes JP, Vergara-Irigaray N et al. . Systemic inflammation induces axon injury during brain inflammation. Ann Neurol 2011;70:932–42. 10.1002/ana.22550
    1. Eisen DP. Manifold beneficial effects of acetyl salicylic acid and nonsteroidal anti-inflammatory drugs on sepsis. Intensive Care Med 2012;38:1249–57. 10.1007/s00134-012-2570-8
    1. Popiolek-Barczyk K, Makuch W, Rojewska E et al. . Inhibition of intracellular signaling pathways NF-kappaB and MEK1/2 attenuates neuropathic pain development and enhances morphine analgesia. Pharmacol Rep 2014;66:845–51. 10.1016/j.pharep.2014.05.001
    1. Hu SS, Bradshaw HB, Chen JS et al. . Prostaglandin E2 glycerol ester, an endogenous COX-2 metabolite of 2-arachidonoylglycerol, induces hyperalgesia and modulates NFkappaB activity. Br J Pharmacol 2008;153:1538–49. 10.1038/bjp.2008.33
    1. Mika J. Modulation of microglia can attenuate neuropathic pain symptoms and enhance morphine effectiveness. Pharmacol Rep 2008;60:297–307.
    1. Kanda H, Kobayashi K, Yamanaka H et al. . COX-1-dependent prostaglandin D2 in microglia contributes to neuropathic pain via DP2 receptor in spinal neurons. Glia 2013;61:943–56. 10.1002/glia.22487
    1. Tatsumi E, Yamanaka H, Kobayashi K et al. . RhoA/ROCK pathway mediates p38 MAPK activation and morphological changes downstream of P2Y12/13 receptors in spinal microglia in neuropathic pain. Glia 2015;63:216–28. 10.1002/glia.22745
    1. Vo T, Rice AS, Dworkin RH. Non-steroidal anti-inflammatory drugs for neuropathic pain: how do we explain continued widespread use? Pain 2009;143:169–71. 10.1016/j.pain.2009.03.013
    1. Genet F, Kulina I, Vaquette C et al. . Neurological heterotopic ossification following spinal cord injury is triggered by macrophage-mediated inflammation in muscle. J Pathol 2015;236:229–40. 10.1002/path.4519
    1. Banovac K, Williams JM, Patrick LD et al. . Prevention of heterotopic ossification after spinal cord injury with indomethacin. Spinal Cord 2001;39:370–4. 10.1038/sj.sc.3101166
    1. van Kuijk AA, Geurts AC, van Kuppevelt HJ. Neurogenic heterotopic ossification in spinal cord injury. Spinal Cord 2002;40:313–26. 10.1038/sj.sc.3101309
    1. Hirschberg DL, Yoles E, Belkin M et al. . Inflammation after axonal injury has conflicting consequences for recovery of function: rescue of spared axons is impaired but regeneration is supported. J Neuroimmunol 1994;50:9–16. 10.1016/0165-5728(94)90209-7
    1. Guth L, Zhang Z, Roberts E. Key role for pregnenolone in combination therapy that promotes recovery after spinal cord injury. Proc Natl Acad Sci USA 1994;91:12308–12. 10.1073/pnas.91.25.12308
    1. Pantovic R, Draganic P, Erakovic V et al. . Effect of indomethacin on motor activity and spinal cord free fatty acid content after experimental spinal cord injury in rabbits. Spinal Cord 2005;43:519–26. 10.1038/sj.sc.3101763
    1. Redondo-Castro E, Navarro X. Chronic ibuprofen administration reduces neuropathic pain but does not exert neuroprotection after spinal cord injury in adult rats. Exp Neurol 2014;252: 95–103. 10.1016/j.expneurol.2013.11.008
    1. Ioannidis JP. Why most published research findings are false. PLoS Med 2005;2:e124 10.1371/journal.pmed.0020124
    1. Strittmatter SM. Overcoming drug development bottlenecks with repurposing: old drugs learn new tricks. Nat Med 2014;20:590–1. 10.1038/nm.3595
    1. Bardou M, Quenot JP, Barkun A. Stress-related mucosal disease in the critically ill patient. Nat Rev Gastroenterol Hepatol 2015;12:98–107. 10.1038/nrgastro.2014.235
    1. Kewalramani LS. Neurogenic gastroduodenal ulceration and bleeding associated with spinal cord injuries. J Trauma 1979;19:259–65. 10.1097/00005373-197904000-00008
    1. Vesterinen HM, Sena ES, Egan KJ et al. . Meta-analysis of data from animal studies: a practical guide. J Neurosci Methods 2014;221:92–102. 10.1016/j.jneumeth.2013.09.010
    1. Sena ES, Briscoe CL, Howells DW et al. . Factors affecting the apparent efficacy and safety of tissue plasminogen activator in thrombotic occlusion models of stroke: systematic review and meta-analysis. J Cereb Blood Flow Metab 2010;30:1905–13. 10.1038/jcbfm.20100.116
    1. Basso DM, Beattie MS, Bresnahan JC. A sensitive and reliable locomotor rating scale for open field testing in rats. J Neurotrauma 1995;12:1–21. 10.1089/neu.1995.12.1
    1. Basso DM, Fisher LC, Anderson AJ et al. . Basso Mouse Scale for locomotion detects differences in recovery after spinal cord injury in five common mouse strains. J Neurotrauma 2006;23:635–59. 10.1089/neu.2006.23.635
    1. Wrathall JR, Pettegrew RK, Harvey F. Spinal cord contusion in the rat: production of graded, reproducible, injury groups. Exp Neurol 1985;88:108–22. 10.1016/0014-4886(85)90117-7
    1. Metz GA, Curt A, van de Meent H et al. . Validation of the weight-drop contusion model in rats: a comparative study of human spinal cord injury. J Neurotrauma 2000;17:1–17. 10.1089/neu.2000.17.1
    1. Lammertse D, Tuszynski MH, Steeves JD et al. . Guidelines for the conduct of clinical trials for spinal cord injury as developed by the ICCP panel: clinical trial design. Spinal Cord 2007;45:232–42. 10.1038/sj.sc.3102010
    1. Steeves JD, Lammertse D, Curt A et al. . Guidelines for the conduct of clinical trials for spinal cord injury (SCI) as developed by the ICCP panel: clinical trial outcome measures. Spinal Cord 2007;45:206–21. 10.1038/sj.sc.3102008
    1. Geisslinger G, Dietzel K, Bezler H et al. . Therapeutically relevant differences in the pharmacokinetical and pharmaceutical behavior of ibuprofen lysinate as compared to ibuprofen acid. Int J Clin Pharmacol Ther Toxicol 1989;27:324–8.
    1. Seidl RO, Nusser-Muller-Busch R, Kurzweil M et al. . Dysphagia in acute tetraplegics: a retrospective study. Spinal Cord 2010;48:197–201. 10.1038/sc.2009.102
    1. Center for Drug Evaluation and Research. Guidance for industry, estimating the maximum safe starting dose in initial clinical trials for therapeutics in adult health volunteers 2005.
    1. Mandula H, Parepally JM, Feng R et al. . Role of site-specific binding to plasma albumin in drug availability to brain. J Pharmacol Exp Ther 2006;317:667–75. 10.1124/jpet.105.097402
    1. Mills RF, Adams SS, Cliffe EE et al. . The metabolism of ibuprofen. Xenobiotica 1973;3:589–98. 10.3109/00498257309151547
    1. International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use. ICH Harmonised Tripartite Guideline, Clinical Safety Data Management: Definitions and Standards for Expedited Reporting E2A, 1994.
    1. Galer BS, Jensen MP. Development and preliminary validation of a pain measure specific to neuropathic pain: the Neuropathic Pain Scale. Neurology 1997;48:332–8. 10.1212/WNL.48.2.332
    1. Jensen MP, Dworkin RH, Gammaitoni AR et al. . Assessment of pain quality in chronic neuropathic and nociceptive pain clinical trials with the Neuropathic Pain Scale. J Pain 2005;6:98–106. 10.1016/j.jpain.2004.11.002
    1. Haas BM, Bergstrom E, Jamous A et al. . The inter rater reliability of the original and of the modified Ashworth scale for the assessment of spasticity in patients with spinal cord injury. Spinal Cord 1996;34:560–4. 10.1038/sc.1996.100
    1. Bannwarth B, Lapicque F, Pehourcq F et al. . Stereoselective disposition of ibuprofen enantiomers in human cerebrospinal fluid. Br J Clin Pharmacol 1995;40:266–9. 10.1111/j.1365-2125.1995.tb05783.x
    1. Kirshblum SC, Waring W, Biering-Sorensen F et al. . Reference for the 2011 revision of the International Standards for Neurological Classification of Spinal Cord Injury. J Spinal Cord Med 2011;34:547–54. 10.1179/107902611X13186000420242
    1. Krauss H, Maier D, Buhren V et al. . Development of heterotopic ossifications, blood markers and outcome after radiation therapy in spinal cord injured patients. Spinal Cord 2015;53:345–8. 10.1038/sc.2014.186
    1. Thaller J, Walker M, Kline AJ et al. . The effect of nonsteroidal anti-inflammatory agents on spinal fusion. Orthopedics 2005;28:299–303; quiz 04–5.
    1. Sucato DJ, Lovejoy JF, Agrawal S et al. . Postoperative ketorolac does not predispose to pseudoarthrosis following posterior spinal fusion and instrumentation for adolescent idiopathic scoliosis. Spine (Phila Pa 1976) 2008;33:1119–24. 10.1097/BRS.0b013e31816f6a2a
    1. Lumawig JM, Yamazaki A, Watanabe K. Dose-dependent inhibition of diclofenac sodium on posterior lumbar interbody fusion rates. Spine J 2009;9:343–9. 10.1016/j.spinee.2008.06.455
    1. Tuszynski MH, Steeves JD, Fawcett JW et al. . Guidelines for the conduct of clinical trials for spinal cord injury as developed by the ICCP Panel: clinical trial inclusion/exclusion criteria and ethics. Spinal Cord 2007;45:222–31. 10.1038/sj.sc.3102009
    1. Previnaire JG, Soler JM, El Masri W et al. . Assessment of the sympathetic level of lesion in patients with spinal cord injury. Spinal Cord 2009;47:122–7. 10.1038/sc.2008.87
    1. European Medicines Agency (EMA). Updated advice on use of high-dose ibuprofen 2015 [updated 22 May 2015.
    1. Paulus HE. FDA arthritis advisory committee meeting: risks of agranulocytosis/aplastic anemia, flank pain, and adverse gastrointestinal effects with the use of nonsteroidal antiinflammatory drugs. Arthritis Rheum 1987;30:593–5. 10.1002/art.1780300518
    1. Singh G, Rosen Ramey D. NSAID induced gastrointestinal complications: the ARAMIS perspective—1997. Arthritis, Rheumatism, and Aging Medical Information System. J Rheumatol Suppl 1998;51:8–16.
    1. Lands LC, Stanojevic S. Oral non-steroidal anti-inflammatory drug therapy for cystic fibrosis. Cochrane Database Syst Rev 2007;(4):CD001505 10.1002/14651858.CD001505.pub2
    1. Konstan MW, Hoppel CL, Chai BL et al. . Ibuprofen in children with cystic fibrosis: pharmacokinetics and adverse effects. J Pediatr 1991;118:956–64. 10.1016/S0022-3476(05)82218-8
    1. Pannu N, Nadim MK. An overview of drug-induced acute kidney injury. Crit Care Med 2008;36(4 Suppl):S216–23. 10.1097/CCM.0b013e318168e375
    1. Whelton A. Nephrotoxicity of nonsteroidal anti-inflammatory drugs: physiologic foundations and clinical implications. Am J Med 1999;106:13S–24S. 10.1016/S0002-9343(99)00113-8
    1. Navarro-Xavier RA, Newson J, Silveira VL et al. . A new strategy for the identification of novel molecules with targeted proresolution of inflammation properties. J Immunol 2010;184:1516–25. 10.4049/jimmunol.0902866
    1. Kramer JL, Lammertse DP, Schubert M et al. . Relationship between motor recovery and independence after sensorimotor-complete cervical spinal cord injury. Neurorehabil Neural Repair 2012;26:1064–71. 10.1177/1545968312447306
    1. Shavelle RM, DeVivo MJ, Brooks JC et al. . Improvements in long-term survival after spinal cord injury? Arch Phys Med Rehabil 2015;96:645–51. 10.1016/j.apmr.2014.11.003

Source: PubMed

3
Se inscrever