Postoperative changes in cognition and cerebrospinal fluid neurodegenerative disease biomarkers

Miles Berger, Jeffrey N Browndyke, Mary Cooter Wright, Chloe Nobuhara, Melody Reese, Leah Acker, W Michael Bullock, Brian J Colin, Michael J Devinney, Eugene W Moretti, Judd W Moul, Brian Ohlendorf, Daniel T Laskowitz, Teresa Waligorska, Leslie M Shaw, Heather E Whitson, Harvey J Cohen, Joseph P Mathew, MADCO-PC Investigators, C L Amundsen, E Bennett, D G Blazer, M P Bolognesi, R Brassard, B E Brigman, J Carter, J Chapman, V Cheong, T A D'Amico, J K DeOrio, D Erdmann, R M Esclamado, M N Ferrandino, B Funk, J C Gadsden, S Grant, G E Garrigues, J R Guercio, A S Habib, R K Hallows, D H Harpole, M G Hartwig, S T Hollenbeck, J Hu, E Iboaya, B A Inman, D W Jang, J Kaisen, A Khan, S Lagoo-Deenadayalan, P S Lee, W T Lee, J Lemm, H Levinson, M E Lipkin, C R Mantyh, K T Martucci, D L McDonagh, J Migaly, S K Mithani, P Mosca, M F Newman, K Ni, T Novick, T N Pappas, A N Perez, A C Peterson, T J Polascik, A Podgoreanu, G M Preminger, Q J Quinones, E N Rampersaud, K Roberts, C N Robertson, S Roman, S Runyon, A Sandler, C D Scales, R P Scheri, S K Smith, L Talbot, J K M Thacker, J Thomas, B C Tong, Y Toulgoat-Dubois, A Tu, S N Vaslef, N Waldron, D S Warner, X Wang, S S Wellman, T Wickenheisser, M Woldorff, C Young, S Zani, Miles Berger, Jeffrey N Browndyke, Mary Cooter Wright, Chloe Nobuhara, Melody Reese, Leah Acker, W Michael Bullock, Brian J Colin, Michael J Devinney, Eugene W Moretti, Judd W Moul, Brian Ohlendorf, Daniel T Laskowitz, Teresa Waligorska, Leslie M Shaw, Heather E Whitson, Harvey J Cohen, Joseph P Mathew, MADCO-PC Investigators, C L Amundsen, E Bennett, D G Blazer, M P Bolognesi, R Brassard, B E Brigman, J Carter, J Chapman, V Cheong, T A D'Amico, J K DeOrio, D Erdmann, R M Esclamado, M N Ferrandino, B Funk, J C Gadsden, S Grant, G E Garrigues, J R Guercio, A S Habib, R K Hallows, D H Harpole, M G Hartwig, S T Hollenbeck, J Hu, E Iboaya, B A Inman, D W Jang, J Kaisen, A Khan, S Lagoo-Deenadayalan, P S Lee, W T Lee, J Lemm, H Levinson, M E Lipkin, C R Mantyh, K T Martucci, D L McDonagh, J Migaly, S K Mithani, P Mosca, M F Newman, K Ni, T Novick, T N Pappas, A N Perez, A C Peterson, T J Polascik, A Podgoreanu, G M Preminger, Q J Quinones, E N Rampersaud, K Roberts, C N Robertson, S Roman, S Runyon, A Sandler, C D Scales, R P Scheri, S K Smith, L Talbot, J K M Thacker, J Thomas, B C Tong, Y Toulgoat-Dubois, A Tu, S N Vaslef, N Waldron, D S Warner, X Wang, S S Wellman, T Wickenheisser, M Woldorff, C Young, S Zani

Abstract

Objective: Numerous investigators have theorized that postoperative changes in Alzheimer's disease neuropathology may underlie postoperative neurocognitive disorders. Thus, we determined the relationship between postoperative changes in cognition and cerebrospinal (CSF) tau, p-tau-181p, or Aβ levels after non-cardiac, non-neurologic surgery in older adults.

Methods: Participants underwent cognitive testing before and 6 weeks after surgery, and lumbar punctures before, 24 h after, and 6 weeks after surgery. Cognitive scores were combined via factor analysis into an overall cognitive index. In total, 110 patients returned for 6-week postoperative testing and were included in the analysis.

Results: There was no significant change from before to 24 h or 6 weeks following surgery in CSF tau (median [median absolute deviation] change before to 24 h: 0.00 [4.36] pg/mL, p = 0.853; change before to 6 weeks: -1.21 [3.98] pg/mL, p = 0.827). There were also no significant changes in CSF p-tau-181p or Aβ over this period. There was no change in cognitive index (mean [95% CI] 0.040 [-0.018, 0.098], p = 0.175) from before to 6 weeks after surgery, although there were postoperative declines in verbal memory (-0.346 [-0.523, -0.170], p = 0.003) and improvements in executive function (0.394, [0.310, 0.479], p < 0.001). There were no significant correlations between preoperative to 6-week postoperative changes in cognition and CSF tau, p-tau-181p, or Aβ42 changes over this interval (p > 0.05 for each).

Interpretation: Neurocognitive changes after non-cardiac, non-neurologic surgery in the majority of cognitively healthy, community-dwelling older adults are unlikely to be related to postoperative changes in AD neuropathology (as assessed by CSF Aβ, tau or p-tau-181p levels or the p-tau-181p/Aβ or tau/Aβ ratios).

Trial registration: clinicaltrials.gov (NCT01993836).

Conflict of interest statement

M. B. has received material support (i.e., EEG monitor loan) for a postoperative recovery study in older adults from Masimo, and legal consulting fees related to postoperative neurocognitive function in older adults. J. N. B. acknowledges funding from Claret Medical, Inc. D. T. L. is an officer of AegisCN, which is developing the ApoE mimetic peptide CN‐105 for clinical use. The other authors have no other conflicts of interest to disclose related to this manuscript.

© 2022 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.

Figures

Figure 1
Figure 1
MADCO‐PC study flow diagram. Out of 140 patients who consented to and enrolled in the study and underwent baseline testing, 110 patients returned for cognitive testing at 6 weeks after surgery. These 110 patients were used in the final data analysis.
Figure 2
Figure 2
CSF biomarkers in MADCO‐PC surgical patients before, 24 h after, and 6 weeks after anesthesia and surgery. In (A–F) black lines represent the group medians, error bars represent MADs. Colored lines in (A–E) represent biomarker trajectories for individual patients who had values >1.5 the interquartile range from the median (a standard statistical definition for outliers 75 ) at any time point. (A) CSF Aβ levels. CSF Aβ42 levels were within 1.5 times the IQR from the group median at all three time points in all patients. (B) CSF tau levels. Six patients had CSF tau levels >1.5 times the IQR from the group median at one or more time points. (C) CSF p‐tau‐181p levels. Nine patients had CSF p‐tau‐181p levels >1.5 times the IQR from the group median at one or more time points. In one of the two patients with p‐tau‐181p levels >90 at the 6‐week time point, CSF samples were unavailable at the preoperative and 24 h time points, so these values were imputed (see Methods section). (D) CSF tau/Aβ ratio. Sixteen patients had CSF tau/Aβ42 ratios >1.5 times the IQR from the group median at one or more time points. (E) CSF p‐tau‐181p/Aβ ratio. 14 patients had CSF p‐tau‐181p/Aβ42 ratios >1.5 times the IQR from the group median at one or more time points. In one of the two patients with a p‐tau‐181p/Aβ ratio >0.5 at the 6‐week time point, CSF samples were unavailable at the preoperative and 24 h time points, so these values were imputed (see Methods section). (F) CSF tau versus Aβ plot in MADCO‐PC patients, with dashed lines representing the cutoffs for pathological Aβ and tau levels. [Colour figure can be viewed at wileyonlinelibrary.com]
Figure 3
Figure 3
Cognitive results before and 6 weeks after surgery. (A) overall cognitive index; (B) verbal memory; (C) visual memory; (D) executive function; (E) attention/concentration. A positive score on the y‐axis represents an improvement in cognitive score from before to after surgery, and vice versa. * indicates a significant difference between the preoperative and 6‐week timepoints at p < 0.05.
Figure 4
Figure 4
Correlation between changes in CSF biomarkers and overall cognitive index change from before to 6 weeks after surgery and anesthesia. (A) CSF Aβ; (B) CSF tau; (C) CSF p‐tau‐181p; (D) tau/Aβ ratio; (E) p‐tau‐181p/Aβ ratio. A positive score on the x‐ or y‐axis represents an improvement in cognitive score or an increase in the indicated biomarker from before to after surgery, respectively, and vice versa.

References

    1. Yang R, Wolfson M, Lewis MC. Unique aspects of the elderly surgical population: an anesthesiologist's perspective. Geriatr Orthop Surg Rehabil. 2011;2(2):56‐64.
    1. 2021 Alzheimer's disease facts and figures. Alzheimers Dement. 2021;17(3):327‐406.
    1. Chen CW, Lin CC, Chen KB, Kuo YC, Li CY, Chung CJ. Increased risk of dementia in people with previous exposure to general anesthesia: a nationwide population‐based case‐control study. Alzheimers Dement. 2014;10(2):196‐204.
    1. Chen PL, Yang CW, Tseng YK, et al. Risk of dementia after anaesthesia and surgery. Br J Psychiatry. 2014;204(3):188‐193.
    1. Avidan MS, Searleman AC, Storandt M, et al. Long‐term cognitive decline in older subjects was not attributable to noncardiac surgery or major illness. Anesthesiology. 2009;111(5):964‐970.
    1. Fischer P, Wallner H, Jungwirth S, et al. Cumulative exposure to general anesthesias and cognitive dysfunction at age 75 in the Vienna Transdanube Aging "VITA" study. J Neuropsychiatry Clin Neurosci. 2007;19(1):21‐26.
    1. Sprung J, Jankowski CJ, Roberts RO, et al. Anesthesia and incident dementia: a population‐based, nested, case‐control study. Mayo Clin Proc. 2013;88(6):552‐561.
    1. Gasparini M, Vanacore N, Schiaffini C, et al. A case‐control study on Alzheimer's disease and exposure to anesthesia. Neurol Sci. 2002;23(1):11‐14.
    1. Harmanci H, Emre M, Gurvit H, et al. Risk factors for Alzheimer disease: a population‐based case‐control study in Istanbul, Turkey. Alzheimer Dis Assoc Disord. 2003;17(3):139‐145.
    1. Evered L, Silbert B, Scott DA, Ames D, Maruff P, Blennow K. Cerebrospinal fluid biomarker for Alzheimer disease predicts postoperative cognitive dysfunction. Anesthesiology. 2016;124(2):353‐361.
    1. Evered L, Silbert B, Knopman DS, et al. Recommendations for the nomenclature of cognitive change associated with anaesthesia and surgery‐2018. Br J Anaesth. 2018;121(5):1005‐1012.
    1. Newman MF, Kirchner JL, Phillips‐Bute B, et al. Longitudinal assessment of neurocognitive function after coronary‐artery bypass surgery. N Engl J Med. 2001;344(6):395‐402.
    1. Inouye SK, Marcantonio ER, Kosar CM, et al. The short‐term and long‐term relationship between delirium and cognitive trajectory in older surgical patients. Alzheimers Dement. 2016;12(7):766‐775.
    1. Selnes OA, Grega MA, Bailey MM, et al. Cognition 6 years after surgical or medical therapy for coronary artery disease. Ann Neurol. 2008;63(5):581‐590.
    1. Selnes OA, Grega MA, Bailey MM, et al. Do management strategies for coronary artery disease influence 6‐year cognitive outcomes? Ann Thorac Surg. 2009;88(2):445‐454.
    1. Browndyke JN, Berger M, Harshbarger TB, et al. Resting‐state functional connectivity and cognition after major cardiac surgery in older adults without preoperative cognitive impairment: preliminary findings. J Am Geriatr Soc. 2017;65(1):e6‐e12.
    1. Browndyke JN, Berger M, Smith PJ, et al. Task‐related changes in degree centrality and local coherence of the posterior cingulate cortex after major cardiac surgery in older adults. Hum Brain Mapp. 2018;39(2):985‐1003.
    1. Rombouts SA, Barkhof F, Goekoop R, Stam CJ, Scheltens P. Altered resting state networks in mild cognitive impairment and mild Alzheimer's disease: an fMRI study. Hum Brain Mapp. 2005;26(4):231‐239.
    1. Sprung J, Kruthiventi SC, Warner DO, et al. Exposure to surgery under general anaesthesia and brain magnetic resonance imaging changes in older adults. Br J Anaesth. 2019;123(6):808‐817.
    1. Kline RP, Pirraglia E, Cheng H, et al. Surgery and brain atrophy in cognitively normal elderly subjects and subjects diagnosed with mild cognitive impairment. Anesthesiology. 2012;116(3):603‐612.
    1. Eckenhoff RG, Johansson JS, Wei H, et al. Inhaled anesthetic enhancement of amyloid‐beta oligomerization and cytotoxicity. Anesthesiology. 2004;101(3):703‐709.
    1. Xie Z, Culley DJ, Dong Y, et al. The common inhalation anesthetic isoflurane induces caspase activation and increases amyloid beta‐protein level in vivo. Ann Neurol. 2008;64(6):618‐627.
    1. Le Freche H, Brouillette J, Fernandez‐Gomez FJ, et al. Tau phosphorylation and sevoflurane anesthesia: an association to postoperative cognitive impairment. Anesthesiology. 2012;116(4):779‐787.
    1. Planel E, Bretteville A, Liu L, et al. Acceleration and persistence of neurofibrillary pathology in a mouse model of tauopathy following anesthesia. FASEB J. 2009;23(8):2595‐2604.
    1. Whittington RA, Virág L, Gratuze M, et al. Dexmedetomidine increases tau phosphorylation under normothermic conditions in vivo and in vitro. Neurobiol Aging. 2015;36(8):2414‐2428.
    1. Whittington RA, Virág L, Marcouiller F, et al. Propofol directly increases tau phosphorylation. PLoS One. 2011;6(1):e16648.
    1. Wan Y, Xu J, Meng F, et al. Cognitive decline following major surgery is associated with gliosis, beta‐amyloid accumulation, and tau phosphorylation in old mice. Crit Care Med. 2010;38(11):2190‐2198.
    1. Kamer AR, Galoyan SM, Haile M, et al. Meloxicam improves object recognition memory and modulates glial activation after splenectomy in mice. Eur J Anaesthesiol. 2012;29(7):332‐337.
    1. Tang JX, Baranov D, Hammond M, Shaw LM, Eckenhoff MF, Eckenhoff RG. Human Alzheimer and inflammation biomarkers after anesthesia and surgery. Anesthesiology. 2011;115(4):727‐732.
    1. Berger M, Nadler JW, Friedman A, et al. The effect of propofol versus isoflurane anesthesia on human cerebrospinal fluid markers of Alzheimer's disease: results of a randomized trial. J Alzheimers Dis. 2016;52(4):1299‐1310.
    1. Palotas A, Reis HJ, Bogats G, et al. Coronary artery bypass surgery provokes Alzheimer's disease‐like changes in the cerebrospinal fluid. J Alzheimers Dis. 2010;21(4):1153‐1164.
    1. Reinsfelt B, Ricksten SE, Zetterberg H, Blennow K, Freden‐Lindqvist J, Westerlind A. Cerebrospinal fluid markers of brain injury, inflammation, and blood‐brain barrier dysfunction in cardiac surgery. Ann Thorac Surg. 2012;94(2):549‐555.
    1. Reinsfelt B, Westerlind A, Blennow K, Zetterberg H, Ricksten SE. Open‐heart surgery increases cerebrospinal fluid levels of Alzheimer‐associated amyloid beta. Acta Anaesthesiol Scand. 2013;57(1):82‐88.
    1. Ferreira D, Perestelo‐Perez L, Westman E, Wahlund LO, Sarria A, Serrano‐Aguilar P. Meta‐review of CSF core biomarkers in Alzheimer's disease: the state‐of‐the‐art after the new revised diagnostic criteria. Front Aging Neurosci. 2014;6:47.
    1. Olsson B, Lautner R, Andreasson U, et al. CSF and blood biomarkers for the diagnosis of Alzheimer's disease: a systematic review and meta‐analysis. Lancet Neurol. 2016;15(7):673‐684.
    1. Horlocker TT, Wedel DJ, Rowlingson JC, et al. Regional anesthesia in the patient receiving antithrombotic or thrombolytic therapy: American Society of Regional Anesthesia and Pain Medicine Evidence‐Based Guidelines (Third Edition). Reg Anesth Pain Med. 2010;35(1):64‐101.
    1. Nobuhara CK, Bullock WM, Bunning T, et al. A protocol to reduce self‐reported pain scores and adverse events following lumbar punctures in older adults. J Neurol. 2020;267(7):2002‐2006.
    1. Shaw LM, Waligorska T, Fields L, et al. Derivation of cutoffs for the Elecsys((R)) amyloid beta (1‐42) assay in Alzheimer's disease. Alzheimers Dement. 2018;10:698‐705.
    1. Yuan Y. Multiple imputation using SAS software. J Stat Softw. 2011;45(6):1‐25.
    1. McDonagh DL, Mathew JP, White WD, et al. Cognitive function after major noncardiac surgery, apolipoprotein E4 genotype, and biomarkers of brain injury. Anesthesiology. 2010;112(4):852‐859.
    1. Benjamini Y, Drai D, Elmer G, Kafkafi N, Golani I. Controlling the false discovery rate in behavior genetics research. Behav Brain Res. 2001;125(1–2):279‐284.
    1. Albert MS, DeKosky ST, Dickson D, et al. The diagnosis of mild cognitive impairment due to Alzheimer's disease: recommendations from the National Institute on Aging‐Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement. 2011;7(3):270‐279.
    1. McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging‐Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement. 2011;7(3):263‐269.
    1. Sperling RA, Aisen PS, Beckett LA, et al. Toward defining the preclinical stages of Alzheimer's disease: recommendations from the National Institute on Aging‐Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement. 2011;7(3):280‐292.
    1. Shaw LM, Vanderstichele H, Knapik‐Czajka M, et al. Cerebrospinal fluid biomarker signature in Alzheimer's disease neuroimaging initiative subjects. Ann Neurol. 2009;65(4):403‐413.
    1. Zhang B, Tian M, Zheng H, et al. Effects of anesthetic isoflurane and desflurane on human cerebrospinal fluid Aβ and τ level. Anesthesiology. 2013;119(1):52‐60.
    1. Ballweg T, White M, Parker M, et al. Association between plasma tau and postoperative delirium incidence and severity: a prospective observational study. Br J Anaesth. 2021;126(2):458‐466.
    1. Danielson M, Wiklund A, Granath F, et al. Association between cerebrospinal fluid biomarkers of neuronal injury or amyloidosis and cognitive decline after major surgery. Br J Anaesth. 2021;126(2):467‐476.
    1. Yaksh TL, Tozier N, Horais KA, et al. Toxicology profile of N‐methyl‐D‐aspartate antagonists delivered by intrathecal infusion in the canine model. Anesthesiology. 2008;108(5):938‐949.
    1. DeLeo JA, Colburn RW, Rickman AJ, Yeager MP. Intrathecal catheterization alone induces neuroimmune activation in the rat. Eur J Pain. 1997;1(2):115‐122.
    1. Olsson M, Ärlig J, Hedner J, Blennow K, Zetterberg H. Repeated lumbar punctures within 3 days may affect CSF biomarker levels. Fluids Barriers CNS. 2019;16(1):37.
    1. Ancelin ML, de Roquefeuil G, Ledesert B, Bonnel F, Cheminal JC, Ritchie K. Exposure to anaesthetic agents, cognitive functioning and depressive symptomatology in the elderly. Br J Psychiatry. 2001;178:360‐366.
    1. Berger M, Murdoch DM, Staats JS, et al. Flow cytometry characterization of cerebrospinal fluid monocytes in patients with postoperative cognitive dysfunction: a pilot study. Anesth Analg. 2019;129(5):e150‐e154.
    1. Berger M, Terrando N, Smith SK, Browndyke JN, Newman MF, Mathew JP. Neurocognitive function after cardiac surgery: from phenotypes to mechanisms. Anesthesiology. 2018;129(4):829‐851.
    1. Devinney MJ, Mathew JP, Berger M. Postoperative delirium and postoperative cognitive dysfunction: two sides of the same coin? Anesthesiology. 2018;129(3):389‐391.
    1. Klinger RY, Cooter M, Bisanar T, et al. Intravenous lidocaine does not improve neurologic outcomes after cardiac surgery: a randomized controlled trial. Anesthesiology. 2019;130(6):958‐970.
    1. Klinger RY, James OG, Borges‐Neto S, et al. 18F‐florbetapir positron emission tomography‐determined cerebral beta‐amyloid deposition and neurocognitive performance after cardiac surgery. Anesthesiology. 2018;128(4):728‐744.
    1. Saczynski JS, Marcantonio ER, Quach L, et al. Cognitive trajectories after postoperative delirium. N Engl J Med. 2012;367(1):30‐39.
    1. VanDusen KW, Li YJ, Cai V, et al. Cerebrospinal fluid proteome changes in older non‐cardiac surgical patients with postoperative cognitive dysfunction. J Alzheimers Dis. 2021;80(3):1281‐1297.
    1. Mathew JP, Mackensen GB, Phillips‐Bute B, et al. Randomized, double‐blinded, placebo controlled study of neuroprotection with lidocaine in cardiac surgery. Stroke. 2009;40(3):880‐887.
    1. Mathew JP, Mackensen GB, Phillips‐Bute B, et al. Effects of extreme hemodilution during cardiac surgery on cognitive function in the elderly. Anesthesiology. 2007;107(4):577‐584.
    1. Mathew JP, Podgoreanu MV, Grocott HP, et al. Genetic variants in P‐selectin and C‐reactive protein influence susceptibility to cognitive decline after cardiac surgery. J Am Coll Cardiol. 2007;49(19):1934‐1942.
    1. Mathew JP, White WD, Schinderle DB, et al. Intraoperative magnesium administration does not improve neurocognitive function after cardiac surgery. Stroke. 2013;44(12):3407‐3413.
    1. Messerotti Benvenuti S, Zanatta P, Valfre C, Polesel E, Palomba D. Preliminary evidence for reduced preoperative cerebral blood flow velocity as a risk factor for cognitive decline three months after cardiac surgery: an extension study. Perfusion. 2012;27(6):486‐492.
    1. Moller JT, Cluitmans P, Rasmussen LS, et al. Long‐term postoperative cognitive dysfunction in the elderly ISPOCD1 study. ISPOCD investigators. International Study of Post‐Operative Cognitive Dysfunction. Lancet. 1998;351(9106):857‐861.
    1. Monk TG, Weldon BC, Garvan CW, et al. Predictors of cognitive dysfunction after major noncardiac surgery. Anesthesiology. 2008;108(1):18‐30.
    1. Nadelson MR, Sanders RD, Avidan MS. Perioperative cognitive trajectory in adults. Br J Anaesth. 2014;112(3):440‐451.
    1. Puskas F, Grocott HP, White WD, Mathew JP, Newman MF, Bar‐Yosef S. Intraoperative hyperglycemia and cognitive decline after CABG. Ann Thorac Surg. 2007;84(5):1467‐1473.
    1. Radtke FM, Franck M, Lendner J, Kruger S, Wernecke KD, Spies CD. Monitoring depth of anaesthesia in a randomized trial decreases the rate of postoperative delirium but not postoperative cognitive dysfunction. Br J Anaesth. 2013;110(suppl 1):i98‐i105.
    1. Van Dijk D, Jansen EW, Hijman R, et al. Cognitive outcome after off‐pump and on‐pump coronary artery bypass graft surgery: a randomized trial. JAMA. 2002;287(11):1405‐1412.
    1. Masters CL, Bateman R, Blennow K, Rowe CC, Sperling RA, Cummings JL. Alzheimer's disease. Nat Rev Dis Primers. 2015;1:15056.
    1. Kennedy RE, Cutter GR, Wang G, Schneider LS. Challenging assumptions about African American participation in Alzheimer disease trials. Am J Geriatr Psychiatry. 2017;25(10):1150‐1159.
    1. Mateen FJ. Is it time for quotas to achieve racial and ethnic representation in multiple sclerosis trials? Front Neurol. 2021;12:680912.
    1. Duff K. Evidence‐based indicators of neuropsychological change in the individual patient: relevant concepts and methods. Arch Clin Neuropsychol. 2012;27(3):248‐261.
    1. Rosner B. Fundamentals of Biostatistics. Thomson‐Brooks/Cole; 2006.

Source: PubMed

3
Sottoscrivi