Recent advances in traumatic brain injury

Abdelhakim Khellaf, Danyal Zaman Khan, Adel Helmy, Abdelhakim Khellaf, Danyal Zaman Khan, Adel Helmy

Abstract

Traumatic brain injury (TBI) is the most common cause of death and disability in those aged under 40 years in the UK. Higher rates of morbidity and mortality are seen in low-income and middle-income countries making it a global health challenge. There has been a secular trend towards reduced incidence of severe TBI in the first world, driven by public health interventions such as seatbelt legislation, helmet use, and workplace health and safety regulations. This has paralleled improved outcomes following TBI delivered in a large part by the widespread establishment of specialised neurointensive care. This update will focus on three key areas of advances in TBI management and research in moderate and severe TBI: refining neurointensive care protocolized therapies, the recent evidence base for decompressive craniectomy and novel pharmacological therapies. In each section, we review the developing evidence base as well as exploring future trajectories of TBI research.

Keywords: Critical care; Monitoring; Neuroprotection; Neurosurgery; TBI; Therapy; Traumatic brain injury.

Conflict of interest statement

The authors have no conflicts of interest relating to this manuscript to declare.

Figures

Fig. 1
Fig. 1
Multi-modality monitor in neurocritical care—illustrating cerebral microdialysis, intracranial pressure and brain tissue oxygenation monitoring. The microdialysis catheter allows sampling of the brain extracellular fluid by recovering molecules of interest that diffuse across the catheter tip and are recovered within a microvial
Fig. 2
Fig. 2
Summary of the mechanisms of energy failure in traumatic brain injury that lead to increased brain lactate: pyruvate ratio (LP ratio). The conversion of lactate to pyruvate is an oxygen-independent step, whereas oxidative phosphorylation and the tricarboxylic acid cycle are oxygen-dependent. Of note, reduced cerebral blood flow and increased oxygen extraction fraction, which characterize classical ischemia, are typically not seen in microvascular ischemia. Mitochondrial dysfunction in the TBI context can arise from multiple pathological processes, often concurrently (most common shown). Ca2+ ionized calcium, CBF cerebral blood flow, iNOS inducible nitric oxide synthase, LDH lactate dehydrogenase, NAD+ nicotinamide adenine dinucleotide (oxidised form), NADH nicotinamide adenine dinucleotide (reduced form), NO nitric oxide, O2 oxygen, O2·−, superoxide radical, OH· hydroxyl radical, pO2 tissue oxygen saturation, ROS reactive oxygen species, TCA tricarboxylic acid cycle
Fig. 3
Fig. 3
Bar chart of RESCUE-ICP trial outcomes at 6 months [47]. Outcomes are displayed using the Extended Glasgow Outcome Scale (eGOS) on the horizontal axis. eGOS at 6 months represents the primary outcome measure of this trial. The percentage of patients falling within the respective outcome category is displayed in the figure table and illustrated in the corresponding graph. “Favourable” outcomes were defined as upper severe disability or better in the RESCUE-ICP trial. “Unfavourable” outcomes comprise of lower severe disability and vegetative state
Fig. 4
Fig. 4
a Bifrontal decompressive craniectomy with the dotted line on the dura representing durotomy site and the red line illustrating an area of falxotomy. b Decompressive hemi-craniectomy with the dotted line representing durotomy incision. Adapted with permission from Timofeev et al. (2012) [43]
Fig. 5
Fig. 5
Bar chart of RESCUE-ICP trial outcomes at 6 months (47). Outcomes are displayed using the Extended Glasgow Outcome Scale (eGOS) on the horizontal axis. eGOS at 6 months represents the primary outcome measure of this trial. The percentage of patients falling within the respective outcome category is displayed in the figure table and illustrated in the corresponding graph. “Favourable” outcomes were defined as upper severe disability or better in the RESCUE-ICP trial. “Unfavourable” outcomes comprise of lower severe disability and vegetative state

References

    1. Timofeev I, Santarius T, Kolias AG, Hutchinson PJA. Decompressive craniectomy—operative technique and perioperative care. Advances and technical standards in neurosurgery. New York: Springer; 2012. pp. 115–136.
    1. Faul M, Wald MM, Xu L, Coronado VG (2010) Traumatic brain injury in the United States; emergency department visits, hospitalizations, and deaths, 2002–2006
    1. Maas AIR, Menon DK, Adelson PD, Andelic N, Bell MJ, Belli A, et al. Traumatic brain injury: integrated approaches to improve prevention, clinical care, and research. Lancet Neurol. 2017;16(12):987–1048.
    1. Excellence NIfHCaC (2017) Head injury: assessment and early management—clinical guideline [CG176] 2017. . Accessed 1 July 2019
    1. Huber A, Dorn A, Witzmann A, Cervós-Navarro J. Microthrombi formation after severe head trauma. Int J Leg Med. 1993;106(3):152–155.
    1. Hartings JA, Bullock MR, Okonkwo DO, Murray LS, Murray GD, Fabricius M, et al. Spreading depolarisations and outcome after traumatic brain injury: a prospective observational study. Lancet Neurol. 2011;10(12):1058–1064.
    1. Teasdale G, Jennett B. Assessment of coma and impaired consciousness: a practical scale. Lancet. 1974;304(7872):81–84.
    1. Mena JH, Sanchez AI, Rubiano AM, Peitzman AB, Sperry JL, Gutierrez MI, et al. Effect of the modified Glasgow Coma Scale score criteria for mild traumatic brain injury on mortality prediction: comparing classic and modified Glasgow Coma Scale score model scores of 13. J Trauma. 2011;71(5):1185–1192.
    1. Dikmen SS, Machamer JE, Powell JM, Temkin NR. Outcome 3–5 years after moderate to severe traumatic brain injury. Arch Phys Med Rehabil. 2003;84(10):1449–1457.
    1. Rosenfeld JV, Maas AI, Bragge P, Morganti-Kossmann MC, Manley GT, Gruen RL. Early management of severe traumatic brain injury. Lancet. 2012;380(9847):1088–1098.
    1. Patel HC, Bouamra O, Woodford M, King AT, Yates DW, Lecky FE. Trends in head injury outcome from 1989 to 2003 and the effect of neurosurgical care: an observational study. Lancet (London, England) 2005;366(9496):1538–1544.
    1. Chesnut RM, Marshall LF, Klauber MR, Blunt BA, Baldwin N, Eisenberg HM, et al. The role of secondary brain injury in determining outcome from severe head injury. J Trauma. 1993;34(2):216–222.
    1. Carney N, Totten AM, O’Reilly C, Ullman JS, Hawryluk GWJ, Bell MJ, et al. Guidelines for the management of severe traumatic brain injury. Neurosurgery. 2017;80(1):6–15.
    1. Chesnut RM, Temkin N, Carney N, Dikmen S, Rondina C, Videtta W, et al. A trial of intracranial-pressure monitoring in traumatic brain injury. N Engl J Med. 2012;367(26):2471–2481.
    1. Smielewski P, Lavinio A, Timofeev I, Radolovich D, Perkes I, Pickard JD, et al. ICM+, a flexible platform for investigations of cerebrospinal dynamics in clinical practice. Acta Neurochir Suppl. 2008;102:145–151.
    1. Moberg (2019) CNS Monitor 2019. . Accessed 1 July 2019
    1. Anandic Medical Systems (2019) BedMasterEx 2019. . Accessed 1 July 2019
    1. Okonkwo DO, Shutter LA, Moore C, Temkin NR, Puccio AM, Madden CJ, et al. Brain oxygen optimization in severe traumatic brain injury phase-II: a phase II randomized trial. Crit Care Med. 2017;45(11):1907–1914.
    1. NIH SIREN Emergencies Trials Network (2018) Brain oxygen optimization in severe TBI phase-3 2018. . Accessed 1 July 2019
    1. MDialysis (2018) ISCUSflex Microdialysis Analyzer 2018. . Accessed 1 July 2019
    1. Hutchinson PJ, Jalloh I, Helmy A, Carpenter KL, Rostami E, Bellander BM, et al. Consensus statement from the 2014 International Microdialysis Forum. Intensive Care Med. 2015;41(9):1517–1528.
    1. Oddo M, Schmidt JM, Carrera E, Badjatia N, Connolly ES, Presciutti M, et al. Impact of tight glycemic control on cerebral glucose metabolism after severe brain injury: a microdialysis study. Crit Care Med. 2008;36(12):3233–3238.
    1. Stein NR, McArthur DL, Etchepare M, Vespa PM. Early cerebral metabolic crisis after TBI influences outcome despite adequate hemodynamic resuscitation. Neurocrit Care. 2012;17(1):49–57.
    1. Dizdarevic K, Hamdan A, Omerhodzic I, Kominlija-Smajic E. Modified Lund concept versus cerebral perfusion pressure-targeted therapy: a randomised controlled study in patients with secondary brain ischaemia. Clin Neurol Neurosurg. 2012;114(2):142–148.
    1. Timofeev I, Carpenter KL, Nortje J, Al-Rawi PG, O’Connell MT, Czosnyka M, et al. Cerebral extracellular chemistry and outcome following traumatic brain injury: a microdialysis study of 223 patients. Brain J Neurol. 2011;134(Pt 2):484–494.
    1. Vespa PM, McArthur D, O’Phelan K, Glenn T, Etchepare M, Kelly D, et al. Persistently low extracellular glucose correlates with poor outcome 6 months after human traumatic brain injury despite a lack of increased lactate: a microdialysis study. J Cereb Blood Flow Metab. 2003;23(7):865–877.
    1. Menon DK, Coles JP, Gupta AK, Fryer TD, Smielewski P, Chatfield DA, et al. Diffusion limited oxygen delivery following head injury. Crit Care Med. 2004;32(6):1384–1390.
    1. Zeiler FA, Thelin EP, Helmy A, Czosnyka M, Hutchinson PJA, Menon DK. A systematic review of cerebral microdialysis and outcomes in TBI: relationships to patient functional outcome, neurophysiologic measures, and tissue outcome. Acta Neurochir. 2017;159(12):2245–2273.
    1. Brain Trauma Foundation . Guidelines for the management of severe traumatic brain injury. New York: Mary Ann Liebert Inc Publications; 2016.
    1. Zafar SN, Khan AA, Ghauri AA, Shamim MS. Phenytoin versus leviteracetam for seizure prophylaxis after brain injury—a meta analysis. BMC Neurol. 2012;12:30.
    1. Inaba K, Menaker J, Branco BC, Gooch J, Okoye OT, Herrold J, et al. A prospective multicenter comparison of levetiracetam versus phenytoin for early posttraumatic seizure prophylaxis. J Trauma Acute Care Surg. 2013;74(3):766–771.
    1. Bhullar IS, Johnson D, Paul JP, Kerwin AJ, Tepas JJ, 3rd, Frykberg ER. More harm than good: antiseizure prophylaxis after traumatic brain injury does not decrease seizure rates but may inhibit functional recovery. J Trauma Acute Care Surg. 2014;76(1):54–60.
    1. Depreitere B, Guiza F, Van den Berghe G, Schuhmann MU, Maier G, Piper I, et al. Pressure autoregulation monitoring and cerebral perfusion pressure target recommendation in patients with severe traumatic brain injury based on minute-by-minute monitoring data. J Neurosurg. 2014;120(6):1451–1457.
    1. Donnelly J, Budohoski KP, Smielewski P, Czosnyka M. Regulation of the cerebral circulation: bedside assessment and clinical implications. Crit Care (London, England) 2016;20(1):129.
    1. Steiner LA, Czosnyka M, Piechnik SK, Smielewski P, Chatfield D, Menon DK, et al. Continuous monitoring of cerebrovascular pressure reactivity allows determination of optimal cerebral perfusion pressure in patients with traumatic brain injury. Crit Care Med. 2002;30(4):733–738.
    1. Sorrentino E, Diedler J, Kasprowicz M, Budohoski KP, Haubrich C, Smielewski P, et al. Critical thresholds for cerebrovascular reactivity after traumatic brain injury. Neurocrit Care. 2012;16(2):258–266.
    1. Undén L, Calcagnile O, Undén J, Reinstrup P, Bazarian J. Validation of the Scandinavian guidelines for initial management of minimal, mild and moderate traumatic brain injury in adults. BMC Med. 2015;13:292.
    1. Thelin EP, Zeiler FA, Ercole A, Mondello S, Buki A, Bellander BM, et al. Serial sampling of serum protein biomarkers for monitoring human traumatic brain injury dynamics: a systematic review. Front Neurol. 2017;8:300.
    1. Choi HA, Badjatia N, Mayer SA. Hypothermia for acute brain injury—mechanisms and practical aspects. Nat Rev Neurol. 2012;8:214.
    1. Andrews PJ, Sinclair HL, Rodriguez A, Harris B, Rhodes J, Watson H, et al. Therapeutic hypothermia to reduce intracranial pressure after traumatic brain injury: the Eurotherm3235 RCT. Health Technol Assess (Winchester, England) 2018;22(45):1–134.
    1. Andrews PJ, Sinclair HL, Rodriguez A, Harris BA, Battison CG, Rhodes JK, et al. Hypothermia for intracranial hypertension after traumatic brain injury. N Engl J Med. 2015;373(25):2403–2412.
    1. Lewis SR, Evans DJ, Butler AR, Schofield-Robinson OJ, Alderson P. Hypothermia for traumatic brain injury. Cochrane Database Syst Re. 2017;9:CD001048.
    1. Cooper DJ, Nichol AD, Bailey M, Bernard S, Cameron PA, Pili-Floury S, et al. Effect of early sustained prophylactic hypothermia on neurologic outcomes among patients with severe traumatic brain injury: the POLAR randomized clinical trial. JAMA. 2018;320(21):2211–2220.
    1. Jiang J-Y, Xu W, Li W-P, Xu W-H, Zhang J, Bao Y-H, et al. Efficacy of standard trauma craniectomy for refractory intracranial hypertension with severe traumatic brain injury: a multicenter, prospective, randomized controlled study. J Neurotrauma. 2005;22(6):623–628.
    1. Kolias AG, Kirkpatrick PJ, Hutchinson PJ. Decompressive craniectomy: past, present and future. Nat Rev Neurol. 2013;9(7):405.
    1. Cooper DJ, Rosenfeld JV, Murray L, Arabi YM, Davies AR, D’Urso P, et al. Decompressive craniectomy in diffuse traumatic brain injury. N Engl J Med. 2011;364(16):1493–1502.
    1. Hutchinson PJ, Kolias AG, Timofeev IS, Corteen EA, Czosnyka M, Timothy J, et al. Trial of decompressive craniectomy for traumatic intracranial hypertension. N Engl J Med. 2016;375(12):1119–1130.
    1. Jennett B, Snoek J, Bond MR, Brooks N. Disability after severe head injury: observations on the use of the Glasgow Outcome Scale. J Neurol Neurosurg Psychiatry. 1981;44(4):285–293.
    1. Kolias AG, Viaroli E, Rubiano AM, Adams H, Khan T, Gupta D, et al. The current status of decompressive craniectomy in traumatic brain injury. Curr Trauma Rep. 2018;4(4):326–332.
    1. Kramer AH, Deis N, Ruddell S, Couillard P, Zygun DA, Doig CJ, et al. Decompressive craniectomy in patients with traumatic brain injury: are the usual indications congruent with those evaluated in clinical trials? Neurocrit Care. 2016;25(1):10–19.
    1. Hutchinson P, Kolias A (2010) Protocol 14PRT/6944: randomised evaluation of surgery with craniectomy for patients undergoing evacuation of acute subdural haematoma (RESCUE-ASDH)—ISRCTN87370545. Lancet.
    1. Bullock MR, Chesnut R, Ghajar J, Gordon D, Hartl R, Newell DW, et al. Surgical management of traumatic brain injury. Neurosurgery. 2006;58(3):16–24.
    1. NIHR Global Health Research Group on Neurotrauma. . Accessed 1 July 2019
    1. Collaborative European NeuroTrauma Effectiveness Research in Traumatic Brain Injury (CENTER-TBI) 2017. . Accessed 1 July 2019
    1. Roberts I, Yates D, Sandercock P, Farrell B, Wasserberg J, Lomas G, et al. Effect of intravenous corticosteroids on death within 14 days in 10,008 adults with clinically significant head injury (MRC CRASH trial): randomised placebo-controlled trial. Lancet (London, England) 2004;364(9442):1321–1328.
    1. Wright DW, Kellermann AL, Hertzberg VS, Clark PL, Frankel M, Goldstein FC, et al. ProTECT: a randomized clinical trial of progesterone for acute traumatic brain injury. Ann Emerg Med. 2007;49(4):391–402.
    1. Xiao G, Wei J, Yan W, Wang W, Lu Z. Improved outcomes from the administration of progesterone for patients with acute severe traumatic brain injury: a randomized controlled trial. Crit Care (London, England) 2008;12(2):R61.
    1. Skolnick BE, Maas AI, Narayan RK, van der Hoop RG, MacAllister T, Ward JD, et al. A clinical trial of progesterone for severe traumatic brain injury. N Engl J Med. 2014;371(26):2467–2476.
    1. Wright DW, Yeatts SD, Silbergleit R, Palesch YY, Hertzberg VS, Frankel M, et al. Very early administration of progesterone for acute traumatic brain injury. N Engl J Med. 2014;371(26):2457–2466.
    1. Coleman T, Brines M. Science review: recombinant human erythropoietin in critical illness: a role beyond anemia? Crit Care (London, England) 2004;8(5):337–341.
    1. Aloizos S, Evodia E, Gourgiotis S, Isaia EC, Seretis C, Baltopoulos GJ. Neuroprotective effects of erythropoietin in patients with severe closed brain injury. Turk Neurosurg. 2015;25(4):552–558.
    1. Li ZM, Xiao YL, Zhu JX, Geng FY, Guo CJ, Chong ZL, et al. Recombinant human erythropoietin improves functional recovery in patients with severe traumatic brain injury: a randomized, double blind and controlled clinical trial. Clin Neurol Neurosurg. 2016;150:80–83.
    1. Lee J, Cho Y, Choi KS, Kim W, Jang BH, Shin H, et al. Efficacy and safety of erythropoietin in patients with traumatic brain injury: a systematic review and meta-analysis. Am J Emerg Med. 2018;37:1101.
    1. Mizoguchi K, Yokoo H, Yoshida M, Tanaka T, Tanaka M. Amantadine increases the extracellular dopamine levels in the striatum by re-uptake inhibition and by N-methyl-d-aspartate antagonism. Brain Res. 1994;662(1–2):255–258.
    1. Giacino JT, Whyte J, Bagiella E, Kalmar K, Childs N, Khademi A, et al. Placebo-controlled trial of amantadine for severe traumatic brain injury. N Engl J Med. 2012;366(9):819–826.
    1. Ghalaenovi H, Fattahi A, Koohpayehzadeh J, Khodadost M, Fatahi N, Taheri M, et al. The effects of amantadine on traumatic brain injury outcome: a double-blind, randomized, controlled, clinical trial. Brain Inj. 2018;32(8):1050–1055.
    1. Hammond FM, Sherer M, Malec JF, Zafonte RD, Dikmen S, Bogner J, et al. Amantadine did not positively impact cognition in chronic traumatic brain injury: a multi-site, randomized, controlled trial. J Neurotrauma. 2018;35(19):2298–2305.
    1. Dewan Y, Komolafe EO, Mejía-Mantilla JH, Perel P, Roberts I, Shakur H. CRASH-3-tranexamic acid for the treatment of significant traumatic brain injury: study protocol for an international randomized, double-blind, placebo-controlled trial. Trials. 2012;13(1):87.
    1. Dunn CJ, Goa KL. Tranexamic acid: a review of its use in surgery and other indications. Drugs. 1999;57(6):1005–1032.
    1. Harhangi BS, Kompanje EJO, Leebeek FWG, Maas AIR. Coagulation disorders after traumatic brain injury. Acta Neurochir. 2008;150(2):165–175.
    1. Gebel JM, Jr, Jauch EC, Brott TG, Khoury J, Sauerbeck L, Salisbury S, et al. Relative edema volume is a predictor of outcome in patients with hyperacute spontaneous intracerebral hemorrhage. Stroke. 2002;33(11):2636–2641.
    1. Williams-Johnson JA, McDonald AH, Strachan GG, Williams EW. Effects of tranexamic acid on death, vascular occlusive events, and blood transfusion in trauma patients with significant haemorrhage (CRASH-2): a randomised, placebo-controlled trial. West Indian Med J. 2010;59(6):612–624.
    1. CRASH-2 Collaborators (Intracranial Bleeding Study) Effect of tranexamic acid in traumatic brain injury: a nested randomised, placebo controlled trial (CRASH-2 Intracranial Bleeding Study) BMJ. 2011;343:d3795.
    1. Tan HB, Wasiak J, Rosenfeld JV, O’Donohoe TJ, Gruen RL. Citicoline (CDP-choline) for traumatic brain injury. Cochrane Database Syst Rev. 2014;8:CD011217.
    1. Zafonte RD, Bagiella E, Ansel BM, Novack TA, Friedewald WT, Hesdorffer DC, et al. Effect of citicoline on functional and cognitive status among patients with traumatic brain injury: citicoline Brain Injury Treatment Trial (COBRIT) JAMA. 2012;308(19):1993–2000.
    1. Pradillo JM, Denes A, Greenhalgh AD, Boutin H, Drake C, McColl BW, et al. Delayed administration of interleukin-1 receptor antagonist reduces ischemic brain damage and inflammation in comorbid rats. J Cereb Blood Flow Metab. 2012;32(9):1810–1819.
    1. Allan SM, Tyrrell PJ, Rothwell NJ. Interleukin-1 and neuronal injury. Nat Rev Immunol. 2005;5(8):629.
    1. Helmy A, Guilfoyle MR, Carpenter KL, Pickard JD, Menon DK, Hutchinson PJ. Recombinant human interleukin-1 receptor antagonist in severe traumatic brain injury: a phase II randomized control trial. J Cereb Blood Flow Metab. 2014;34(5):845–851.
    1. Helmy A (2017) A randomised double blind placebo controlled dose-range study using placebo, 1.5 g and 3.0 g of intravenous recombinant interleukin-1 receptor antagonist (Anakinra) for patients with moderate-to-severe TBI 2017. . Accessed 1 July 2019

Source: PubMed

3
Se inscrever