Tumor Necrosis Factor (TNF) blocking agents are associated with lower risk for Alzheimer's disease in patients with rheumatoid arthritis and psoriasis

Mengshi Zhou, Rong Xu, David C Kaelber, Mark E Gurney, Mengshi Zhou, Rong Xu, David C Kaelber, Mark E Gurney

Abstract

This large, retrospective case-control study of electronic health records from 56 million unique adult patients examined whether or not treatment with a Tumor Necrosis Factor (TNF) blocking agent is associated with lower risk for Alzheimer's disease (AD) in patients with rheumatoid arthritis (RA), psoriasis, and other inflammatory diseases which are mediated in part by TNF and for which a TNF blocker is an approved treatment. The analysis compared the diagnosis of AD as an outcome measure in patients receiving at least one prescription for a TNF blocking agent (etanercept, adalimumab, and infliximab) or for methotrexate. Adjusted odds ratios (AORs) were estimated using the Cochran-Mantel-Haenszel (CMH) method and presented with 95% confidence intervals (CIs) and p-values. RA was associated with a higher risk for AD (Adjusted Odds Ratio (AOR) = 2.06, 95% Confidence Interval: (2.02-2.10), P-value <0.0001) as did psoriasis (AOR = 1.37 (1.31-1.42), P <0.0001), ankylosing spondylitis (AOR = 1.57 (1.39-1.77), P <0.0001), inflammatory bowel disease (AOR = 2.46 (2.33-2.59), P < 0.0001), ulcerative colitis (AOR = 1.82 (1.74-1.91), P <0.0001), and Crohn's disease (AOR = 2.33 (2.22-2.43), P <0.0001). The risk for AD in patients with RA was lower among patients treated with etanercept (AOR = 0.34 (0.25-0.47), P <0.0001), adalimumab (AOR = 0.28 (0.19-0.39), P < 0.0001), or infliximab (AOR = 0.52 (0.39-0.69), P <0.0001). Methotrexate was also associated with a lower risk for AD (AOR = 0.64 (0.61-0.68), P <0.0001), while lower risk was found in patients with a prescription history for both a TNF blocker and methotrexate. Etanercept and adalimumab also were associated with lower risk for AD in patients with psoriasis: AOR = 0.47 (0.30-0.73 and 0.41 (0.20-0.76), respectively. There was no effect of gender or race, while younger patients showed greater benefit from a TNF blocker than did older patients. This study identifies a subset of patients in whom systemic inflammation contributes to risk for AD through a pathological mechanism involving TNF and who therefore may benefit from treatment with a TNF blocking agent.

Conflict of interest statement

M.E.G is an employee of Tetra Therapeutics. This commercial affiliation does not alter the adherence of M.E.G to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. A screen shot of cohort…
Fig 1. A screen shot of cohort identification using the IBM Watson Health Explorys Cohort Discovery fast search tool.
This example selects female White patients with age > 65 years with a diagnosis code for dementia and a prescription for etanercept.
Fig 2. Cumulative age distribution of patients…
Fig 2. Cumulative age distribution of patients with a diagnosis code for rheumatoid arthritis, dementia or Alzheimer’s disease.
Fig 3
Fig 3
a. Adjusted Odds Ratio (AOR) for a diagnosis of Alzheimer’s disease associated with a diagnosis for an inflammatory disease as compared to the non-inflammatory disease group. Abbreviations: RA -rheumatoid arthritis, AS -ankylosing spondylitis, PA -psoriatic arthritis, IBD -inflammatory bowel disease, UC -ulcerative colitis, Crohn’s -Crohn’s disease. b Adjusted Odds Ratios (AORs) for a diagnosis of Alzheimer’s disease associated with a diagnosis for rheumatoid arthritis as compared to the non-inflammatory disease group, further adjusting for BMI, smoking status, or alcohol use. The added covariate is presented in column 2. We present both adjusted and stratified AOR for each covariate.
Fig 4. Adjusted Odds Ratio (AOR) for…
Fig 4. Adjusted Odds Ratio (AOR) for a diagnosis code of dementia associated with a diagnosis for an inflammatory disease as compared to the non-inflammatory disease group.
Abbreviations: RA -rheumatoid arthritis, AS -ankylosing spondylitis, PA -psoriatic arthritis, IBD -inflammatory bowel disease, UC -ulcerative colitis, Crohn’s -Crohn’s disease.
Fig 5. Adjusted Odds Ratio (AOR) showing…
Fig 5. Adjusted Odds Ratio (AOR) showing inverse risk associations between Alzheimer’s disease or a diagnosis of dementia and prescription history of a TNF blocker (exclude certolizumab pegol and golimumab) or methotrexate compared to the no-drug group in patients with a diagnosis of rheumatoid arthritis with a prescription.
The analysis excluded patients with a prescription history of a TNF blocker and methotrexate.
Fig 6. Adjusted Odds Ratio (AOR) showing…
Fig 6. Adjusted Odds Ratio (AOR) showing inverse associations between Alzheimer’s disease or a diagnosis of dementia and prescription history of methotrexate and a TNF blocker compared to the methotrexate and no-TNF blocker group in patients with a diagnosis of rheumatoid arthritis.
Fig 7. Adjusted Odds Ratio (AOR) showing…
Fig 7. Adjusted Odds Ratio (AOR) showing inverse associations between Alzheimer’s disease or a diagnosis of dementia and prescription history of a TNF blocker (exclude certolizumab pegol and golimumab) or methotrexate compared to the no-drug group in patients with a diagnosis of psoriasis.
Fig 8. Adjusted Odds Ratio (AOR) showing…
Fig 8. Adjusted Odds Ratio (AOR) showing inverse associations between Alzheimer’s disease or a diagnosis of dementia and prescription history of a TNF blocker in patients with a diagnosis of psoriasis adjusting for methotrexate prescription status.
Fig 9. Effect of age, gender and…
Fig 9. Effect of age, gender and race on risk for a diagnosis of dementia in rheumatoid arthritis patients treated with etanercept.
Young patients (18–65 years) showed greater benefit than did older patients (over 65 years). The effect of gender and race was not significant.
Fig 10. Crude Odds Ratios (OR) and…
Fig 10. Crude Odds Ratios (OR) and Adjusted Odds Ratios (AOR) based on insurance status (Medicaid versus no-Medicaid, or private versus no-private insurance) in patients with a diagnosis code for dementia and rheumatoid arthritis.
Fig 11. Comparison of the risk for…
Fig 11. Comparison of the risk for a diagnosis of Alzheimer’s disease or dementia in patients with a diagnosis of rheumatoid arthritis or psoriasis with a prescription history for etanercept, adalimumab, or infliximab who were not prescribed methotrexate against the general population.

References

    1. Scheltens P, Blennow K, Breteler MM, de Strooper B, Frisoni GB, Salloway S, et al. Alzheimer's disease. Lancet. 2016;388(10043):505–17. 10.1016/S0140-6736(15)01124-1 .
    1. Jack CR Jr., Bennett DA, Blennow K, Carrillo MC, Dunn B, Haeberlein SB, et al. NIA-AA Research Framework: Toward a biological definition of Alzheimer's disease. Alzheimers Dement. 2018;14(4):535–62. 10.1016/j.jalz.2018.02.018
    1. Hansen DV, Hanson JE, Sheng M. Microglia in Alzheimer's disease. J Cell Biol. 2018;217(2):459–72. 10.1083/jcb.201709069
    1. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science. 2010;330(6005):841–5. 10.1126/science.1194637
    1. Willrich MA, Murray DL, Snyder MR. Tumor necrosis factor inhibitors: clinical utility in autoimmune diseases. Transl Res. 2015;165(2):270–82. 10.1016/j.trsl.2014.09.006 .
    1. Tarkowski E, Andreasen N, Tarkowski A, Blennow K. Intrathecal inflammation precedes development of Alzheimer's disease. J Neurol Neurosurg Psychiatry. 2003;74(9):1200–5. 10.1136/jnnp.74.9.1200
    1. Cheng X, Shen Y, Li R. Targeting TNF: a therapeutic strategy for Alzheimer's disease. Drug Discov Today. 2014. 10.1016/j.drudis.2014.06.029 .
    1. Long H, Zhong G, Wang C, Zhang J, Zhang Y, Luo J, et al. TREM2 Attenuates Abeta1-42-Mediated Neuroinflammation in BV-2 Cells by Downregulating TLR Signaling. Neurochem Res. 2019;44(8):1830–9. 10.1007/s11064-019-02817-1 .
    1. McAlpine FE, Tansey MG. Neuroinflammation and tumor necrosis factor signaling in the pathophysiology of Alzheimer's disease. J Inflamm Res. 2008;1:29–39. 10.2147/jir.s4397
    1. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature. 2017;541(7638):481–7. 10.1038/nature21029
    1. Keffer J, Probert L, Cazlaris H, Georgopoulos S, Kaslaris E, Kioussis D, et al. Transgenic mice expressing human tumour necrosis factor: a predictive genetic model of arthritis. EMBO J. 1991;10(13):4025–31.
    1. Pan W, Kastin AJ. TNFalpha transport across the blood-brain barrier is abolished in receptor knockout mice. Exp Neurol. 2002;174(2):193–200. 10.1006/exnr.2002.7871 .
    1. Chou RC, Kane M, Ghimire S, Gautam S, Gui J. Treatment for Rheumatoid Arthritis and Risk of Alzheimer's Disease: A Nested Case-Control Analysis. CNS Drugs. 2016;30(11):1111–20. 10.1007/s40263-016-0374-z
    1. Kaelber DC, Foster W, Gilder J, Love TE, Jain AK. Patient characteristics associated with venous thromboembolic events: a cohort study using pooled electronic health record data. J Am Med Inform Assoc. 2012;19(6):965–72. 10.1136/amiajnl-2011-000782
    1. Patel VN, Kaelber DC. Using aggregated, de-identified electronic health record data for multivariate pharmacosurveillance: a case study of azathioprine. J Biomed Inform. 2014;52:36–42. 10.1016/j.jbi.2013.10.009
    1. Winhusen T, Theobald J, Kaelber DC, Lewis D. Medical complications associated with substance use disorders in patients with type 2 diabetes and hypertension: electronic health record findings. Addiction. 2019. 10.1111/add.14607 .
    1. Peppel K, Crawford D, Beutler B. A tumor necrosis factor (TNF) receptor-IgG heavy chain chimeric protein as a bivalent antagonist of TNF activity. J Exp Med. 1991;174(6):1483–9. 10.1084/jem.174.6.1483
    1. Jespers LS, Roberts A, Mahler SM, Winter G, Hoogenboom HR. Guiding the selection of human antibodies from phage display repertoires to a single epitope of an antigen. Biotechnology (N Y). 1994;12(9):899–903. 10.1038/nbt0994-899 .
    1. Knight DM, Trinh H, Le J, Siegel S, Shealy D, McDonough M, et al. Construction and initial characterization of a mouse-human chimeric anti-TNF antibody. Mol Immunol. 1993;30(16):1443–53. 10.1016/0161-5890(93)90106-l .
    1. Pardridge WM. Blood-brain barrier delivery. Drug Discov Today. 2007;12(1–2):54–61. Epub 2007/01/03. 10.1016/j.drudis.2006.10.013 .
    1. Lindberg C. The Unified Medical Language System (UMLS) of the National Library of Medicine. J Am Med Rec Assoc. 1990;61(5):40–2. .
    1. Millar J. The Need for a Global Language—SNOMED CT Introduction. Stud Health Technol Inform. 2016;225:683–5. .
    1. Nelson SJ, Zeng K, Kilbourne J, Powell T, Moore R. Normalized names for clinical drugs: RxNorm at 6 years. J Am Med Inform Assoc. 2011;18(4):441–8. 10.1136/amiajnl-2011-000116
    1. Huff SM, Rocha RA, McDonald CJ, De Moor GJ, Fiers T, Bidgood WD Jr., et al. Development of the Logical Observation Identifier Names and Codes (LOINC) vocabulary. J Am Med Inform Assoc. 1998;5(3):276–92. 10.1136/jamia.1998.0050276
    1. Piazza-Gardner AK, Gaffud TJ, Barry AE. The impact of alcohol on Alzheimer's disease: a systematic review. Aging Ment Health. 2013;17(2):133–46. 10.1080/13607863.2012.742488 .
    1. Profenno LA, Porsteinsson AP, Faraone SV. Meta-analysis of Alzheimer's disease risk with obesity, diabetes, and related disorders. Biol Psychiatry. 2010;67(6):505–12. 10.1016/j.biopsych.2009.02.013 .
    1. Baumgart M, Snyder HM, Carrillo MC, Fazio S, Kim H, Johns H. Summary of the evidence on modifiable risk factors for cognitive decline and dementia: A population-based perspective. Alzheimers Dement. 2015;11(6):718–26. 10.1016/j.jalz.2015.05.016 .
    1. Di Giuseppe D, Alfredsson L, Bottai M, Askling J, Wolk A. Long term alcohol intake and risk of rheumatoid arthritis in women: a population based cohort study. BMJ. 2012;345:e4230 10.1136/bmj.e4230
    1. Lu B, Solomon DH, Costenbader KH, Karlson EW. Alcohol consumption and risk of incident rheumatoid arthritis in women: a prospective study. Arthritis Rheumatol. 2014;66(8):1998–2005. 10.1002/art.38634
    1. Qin B, Yang M, Fu H, Ma N, Wei T, Tang Q, et al. Body mass index and the risk of rheumatoid arthritis: a systematic review and dose-response meta-analysis. Arthritis Res Ther. 2015;17:86 10.1186/s13075-015-0601-x
    1. Sugiyama D, Nishimura K, Tamaki K, Tsuji G, Nakazawa T, Morinobu A, et al. Impact of smoking as a risk factor for developing rheumatoid arthritis: a meta-analysis of observational studies. Ann Rheum Dis. 2010;69(1):70–81. 10.1136/ard.2008.096487 .
    1. Klareskog L, Stolt P, Lundberg K, Kallberg H, Bengtsson C, Grunewald J, et al. A new model for an etiology of rheumatoid arthritis: smoking may trigger HLA-DR (shared epitope)-restricted immune reactions to autoantigens modified by citrullination. Arthritis Rheum. 2006;54(1):38–46. 10.1002/art.21575 .
    1. Gilsanz P, Mayeda ER, Glymour MM, Quesenberry CP, Mungas DM, DeCarli C, et al. Female sex, early-onset hypertension, and risk of dementia. Neurology. 2017;89(18):1886–93. 10.1212/WNL.0000000000004602
    1. Kuritz SJ, Landis JR, Koch GG. A general overview of Mantel-Haenszel methods: applications and recent developments. Annu Rev Public Health. 1988;9:123–60. 10.1146/annurev.pu.09.050188.001011 .
    1. Salloway S, Sperling R, Fox NC, Blennow K, Klunk W, Raskind M, et al. Two phase 3 trials of bapineuzumab in mild-to-moderate Alzheimer's disease. N Engl J Med. 2014;370(4):322–33. 10.1056/NEJMoa1304839 .
    1. Doody RS, Thomas RG, Farlow M, Iwatsubo T, Vellas B, Joffe S, et al. Phase 3 trials of solanezumab for mild-to-moderate Alzheimer's disease. N Engl J Med. 2014;370(4):311–21. 10.1056/NEJMoa1312889 .
    1. Alzheimer's A. 2016 Alzheimer's disease facts and figures. Alzheimers Dement. 2016;12(4):459–509. 10.1016/j.jalz.2016.03.001 .
    1. Aletaha D, Smolen JS. Diagnosis and Management of Rheumatoid Arthritis: A Review. JAMA. 2018;320(13):1360–72. 10.1001/jama.2018.13103 .
    1. Favalli EG, Pregnolato F, Biggioggero M, Becciolini A, Penatti AE, Marchesoni A, et al. Twelve-Year Retention Rate of First-Line Tumor Necrosis Factor Inhibitors in Rheumatoid Arthritis: Real-Life Data From a Local Registry. Arthritis Care Res (Hoboken). 2016;68(4):432–9. 10.1002/acr.22788 .
    1. Papadopoulos CG, Gartzonikas IK, Pappa TK, Markatseli TE, Migkos MP, Voulgari PV, et al. Eight-year survival study of first-line tumour necrosis factor alpha inhibitors in rheumatoid arthritis: real-world data from a university centre registry. Rheumatol Adv Pract. 2019;3(1):rkz007 10.1093/rap/rkz007
    1. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer's disease. Lancet Neurol. 2015;14(4):388–405. 10.1016/S1474-4422(15)70016-5
    1. Butchart J, Brook L, Hopkins V, Teeling J, Puntener U, Culliford D, et al. Etanercept in Alzheimer disease: A randomized, placebo-controlled, double-blind, phase 2 trial. Neurology. 2015;84(21):2161–8. 10.1212/WNL.0000000000001617
    1. Peter I, Dubinsky M, Bressman S, Park A, Lu C, Chen N, et al. Anti-Tumor Necrosis Factor Therapy and Incidence of Parkinson Disease Among Patients With Inflammatory Bowel Disease. JAMA Neurol. 2018;75(8):939–46. 10.1001/jamaneurol.2018.0605
    1. Tyring S, Gottlieb A, Papp K, Gordon K, Leonardi C, Wang A, et al. Etanercept and clinical outcomes, fatigue, and depression in psoriasis: double-blind placebo-controlled randomised phase III trial. Lancet. 2006;367(9504):29–35. 10.1016/S0140-6736(05)67763-X .
    1. Gurney ME, D'Amato EC, Burgin AB. Phosphodiesterase-4 (PDE4) Molecular Pharmacology and Alzheimer's Disease. Neurotherapeutics. 2014. 10.1007/s13311-014-0309-7 .
    1. Chakrabarty P, Li A, Ladd TB, Strickland MR, Koller EJ, Burgess JD, et al. TLR5 decoy receptor as a novel anti-amyloid therapeutic for Alzheimer's disease. J Exp Med. 2018;215(9):2247–64. 10.1084/jem.20180484
    1. Probert L. TNF and its receptors in the CNS: The essential, the desirable and the deleterious effects. Neuroscience. 2015;302:2–22. 10.1016/j.neuroscience.2015.06.038 .
    1. Chung WS, Welsh CA, Barres BA, Stevens B. Do glia drive synaptic and cognitive impairment in disease? Nat Neurosci. 2015;18(11):1539–45. 10.1038/nn.4142
    1. Salter MW, Stevens B. Microglia emerge as central players in brain disease. Nat Med. 2017;23(9):1018–27. 10.1038/nm.4397 .
    1. Sims R, van der Lee SJ, Naj AC, Bellenguez C, Badarinarayan N, Jakobsdottir J, et al. Rare coding variants in PLCG2, ABI3, and TREM2 implicate microglial-mediated innate immunity in Alzheimer's disease. Nat Genet. 2017;49(9):1373–84. 10.1038/ng.3916
    1. Yeh FL, Hansen DV, Sheng M. TREM2, Microglia, and Neurodegenerative Diseases. Trends Mol Med. 2017;23(6):512–33. 10.1016/j.molmed.2017.03.008 .
    1. Jiang T, Zhang YD, Chen Q, Gao Q, Zhu XC, Zhou JS, et al. TREM2 modifies microglial phenotype and provides neuroprotection in P301S tau transgenic mice. Neuropharmacology. 2016;105:196–206. 10.1016/j.neuropharm.2016.01.028 .
    1. Bemiller SM, McCray TJ, Allan K, Formica SV, Xu G, Wilson G, et al. TREM2 deficiency exacerbates tau pathology through dysregulated kinase signaling in a mouse model of tauopathy. Mol Neurodegener. 2017;12(1):74 10.1186/s13024-017-0216-6
    1. Gorenjak V, Aldasoro Arguinano AA, Dade S, Stathopoulou MG, Vance DR, Masson C, et al. The polymorphism rs6918289 located in the downstream region of the TREM2 gene is associated with TNF-alpha levels and IMT-F. Sci Rep. 2018;8(1):7160 10.1038/s41598-018-25553-y .
    1. Li P, Zheng Y, Chen X. Drugs for Autoimmune Inflammatory Diseases: From Small Molecule Compounds to Anti-TNF Biologics. Front Pharmacol. 2017;8:460 10.3389/fphar.2017.00460
    1. Jin SL, Conti M. Induction of the cyclic nucleotide phosphodiesterase PDE4B is essential for LPS-activated TNF-alpha responses. Proc Natl Acad Sci U S A. 2002;99(11):7628–33. Epub 2002/05/29. 10.1073/pnas.122041599 .
    1. Ghosh M, Garcia-Castillo D, Aguirre V, Golshani R, Atkins CM, Bramlett HM, et al. Proinflammatory cytokine regulation of cyclic AMP-phosphodiesterase 4 signaling in microglia in vitro and following CNS injury. Glia. 2012. 10.1002/glia.22401 .
    1. Wilson NM, Gurney ME, Dietrich WD, Atkins CM. Therapeutic benefits of phosphodiesterase 4B inhibition after traumatic brain injury. PLoS One. 2017;12(5):e0178013 10.1371/journal.pone.0178013
    1. Vlad SC, Miller DR, Kowall NW, Felson DT. Protective effects of NSAIDs on the development of Alzheimer disease. Neurology. 2008;70(19):1672–7. 10.1212/01.wnl.0000311269.57716.63
    1. Leoutsakos JM, Muthen BO, Breitner JC, Lyketsos CG, Team AR. Effects of non-steroidal anti-inflammatory drug treatments on cognitive decline vary by phase of pre-clinical Alzheimer disease: findings from the randomized controlled Alzheimer's Disease Anti-inflammatory Prevention Trial. Int J Geriatr Psychiatry. 2012;27(4):364–74. 10.1002/gps.2723
    1. Meyer PF, Tremblay-Mercier J, Leoutsakos J, Madjar C, Lafaille-Maignan ME, Savard M, et al. INTREPAD: A randomized trial of naproxen to slow progress of presymptomatic Alzheimer disease. Neurology. 2019;92(18):e2070–e80. 10.1212/WNL.0000000000007232
    1. Reines SA, Block GA, Morris JC, Liu G, Nessly ML, Lines CR, et al. Rofecoxib: no effect on Alzheimer's disease in a 1-year, randomized, blinded, controlled study. Neurology. 2004;62(1):66–71. 10.1212/wnl.62.1.66 .
    1. Page TH, Turner JJ, Brown AC, Timms EM, Inglis JJ, Brennan FM, et al. Nonsteroidal anti-inflammatory drugs increase TNF production in rheumatoid synovial membrane cultures and whole blood. J Immunol. 2010;185(6):3694–701. 10.4049/jimmunol.1000906 .
    1. Aisen PS, Davis KL, Berg JD, Schafer K, Campbell K, Thomas RG, et al. A randomized controlled trial of prednisone in Alzheimer's disease. Alzheimer's Disease Cooperative Study. Neurology. 2000;54(3):588–93. 10.1212/wnl.54.3.588 .

Source: PubMed

3
Sottoscrivi