Lymphopenia and risk of infection and infection-related death in 98,344 individuals from a prospective Danish population-based study

Marie Warny, Jens Helby, Børge Grønne Nordestgaard, Henrik Birgens, Stig Egil Bojesen, Marie Warny, Jens Helby, Børge Grønne Nordestgaard, Henrik Birgens, Stig Egil Bojesen

Abstract

Background: Neutropenia increases the risk of infection, but it is unknown if this also applies to lymphopenia. We therefore tested the hypotheses that lymphopenia is associated with increased risk of infection and infection-related death in the general population.

Methods and findings: Of the invited 220,424 individuals, 99,191 attended examination. We analyzed 98,344 individuals from the Copenhagen General Population Study (Denmark), examined from November 25, 2003, to July 9, 2013, and with available blood lymphocyte count at date of examination. During a median of 6 years of follow-up, they developed 8,401 infections and experienced 1,045 infection-related deaths. Due to the completeness of the Danish civil and health registries, none of the 98,344 individuals were lost to follow-up, and those emigrating (n = 385) or dying (n = 5,636) had their follow-up truncated at the day of emigration or death. At date of examination, mean age was 58 years, and 44,181 (44.9%) were men. Individuals with lymphopenia (lymphocyte count < 1.1 × 109/l, n = 2,352) compared to those with lymphocytes in the reference range (1.1-3.7 × 109/l, n = 93,538) had multivariable-adjusted hazard ratios of 1.41 (95% CI 1.28-1.56) for any infection, 1.31 (1.14-1.52) for pneumonia, 1.44 (1.15-1.79) for skin infection, 1.26 (1.02-1.56) for urinary tract infection, 1.51 (1.21-1.89) for sepsis, 1.38 (1.01-1.88) for diarrheal disease, 2.15 (1.16-3.98) for endocarditis, and 2.26 (1.21-4.24) for other infections. The corresponding hazard ratio for infection-related death was 1.70 (95% CI 1.37-2.10). Analyses were adjusted for age, sex, smoking status, cumulative smoking, alcohol intake, body mass index, plasma C-reactive protein, blood neutrophil count, recent infection, Charlson comorbidity index, autoimmune diseases, medication use, and immunodeficiency/hematologic disease. The findings were robust in all stratified analyses and also when including only events later than 2 years after first examination. However, due to the observational design, the study cannot address questions of causality, and our analyses might theoretically have been affected by residual confounding and reverse causation. In principle, fluctuating lymphocyte counts over time might also have influenced analyses, but lymphocyte counts in 5,181 individuals measured 10 years after first examination showed a regression dilution ratio of 0.68.

Conclusions: Lymphopenia was associated with increased risk of hospitalization with infection and increased risk of infection-related death in the general population. Notably, causality cannot be deduced from our data.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. Flow chart of individuals from…
Fig 1. Flow chart of individuals from the Copenhagen General Population Study.
Fig 2. Regression toward the mean of…
Fig 2. Regression toward the mean of lymphocyte counts, categorized as lymphopenia (9/l), reference lymphocyte count (1.1–3.7 × 109/l), and lymphocytosis (>3.7 × 109/l) at the date of examination in 2003 in the Copenhagen General Population Study.
When categorizing lymphocyte counts, lymphopenia was defined as a lymphocyte count below the 2.5th percentile, the reference category was defined as a lymphocyte count between the 2.5th and 97.5th percentile, and lymphocytosis was defined as a lymphocyte count above the 97.5th percentile. Median values and interquartile ranges of 5,181 individuals at the 2003 examination (left) and the same individuals—maintaining the 2003 categorization—at the 2013 examination (right). Regression dilution ratio is 2.04/2.98 = 0.68. The table below the graph gives the percentage of individuals in each lymphocyte category at the 2003 examination (rows) and at the 2013 examination (columns).
Fig 3. Risk of any infection as…
Fig 3. Risk of any infection as a function of lymphocyte count for individuals from the Copenhagen General Population Study.
Solid red lines are hazard ratios, and dashed black lines indicate 95% confidence intervals based on fitting of cubic splines to risk estimates obtained using Cox proportional hazards regression. Multivariable adjustment includes all covariates listed in Table 1 except age, but with age as the underlying timescale. The median lymphocyte value of 2.1 × 109/l was set as reference for the continuous model. For the multivariable adjusted model, the lymphocyte interval 30–40 × 109/l is added to illustrate that for very high lymphocyte counts the risk of infections increases, but with a broad confidence interval. When categorizing lymphocyte counts, lymphopenia was defined as a lymphocyte count below the 2.5th percentile, the reference category was defined as a lymphocyte count between the 2.5th and 97.5th percentile, and lymphocytosis was defined as a lymphocyte count above the 97.5th percentile.
Fig 4. Multivariable adjusted risks of specific…
Fig 4. Multivariable adjusted risks of specific infections as a function of lymphocyte count for individuals from the Copenhagen General Population Study.
Solid red lines are multivariable adjusted hazard ratios, and dashed black lines indicate 95% confidence intervals based on fitting of cubic splines to risk estimates obtained using Cox proportional hazards regression. Multivariable adjustment includes all covariates listed in Table 1 except age, but with age as the underlying timescale. The median lymphocyte value of 2.1 × 109/l was set as reference for the continuous model. The sum of the numbers of cases of specific infections exceeds the number of cases of “any infection” since individuals could have more than 1 specific infection. When categorizing lymphocyte counts, lymphopenia was defined as a lymphocyte count below the 2.5th percentile, the reference category was defined as a lymphocyte count between the 2.5th and 97.5th percentile, and lymphocytosis was defined as a lymphocyte count above the 97.5th percentile.
Fig 5. Multivariable adjusted risk of any…
Fig 5. Multivariable adjusted risk of any infection for individuals from the Copenhagen General Population Study with lymphopenia (lymphocyte count 9/l) compared to individuals with lymphocytes in the reference range (1.1–3.7 × 109/l).
Multivariable adjustment includes all covariates listed in Table 1 except age, but with age as the underlying timescale. The sum number of individuals in the strata varies slightly because of varying numbers of individuals with missing data in the covariates. When categorizing lymphocyte counts, lymphopenia was defined as a lymphocyte count below the 2.5th percentile, while the reference category was defined as a lymphocyte count between the 2.5th and 97.5th percentile. NSNot significant after adjusting for 12 multiple comparisons using the Bonferroni method.aAccording to a likelihood ratio test.b≤168 versus >168 g/week for men and ≤84 versus >84 g/week for women.cWhen categorizing neutrophil counts, neutropenia was defined as a neutrophil count below 1.5 × 109/l, which is the generally accepted lower cutoff [,–43], the reference category was defined as a neutrophil count from 1.5 × 109/l to 5.9 × 109/l, and neutrocytosis was defined as a neutrophil count above 5.9 × 109/l.dAs defined by the Charlson comorbidity index.
Fig 6. Multivariable adjusted risk of any…
Fig 6. Multivariable adjusted risk of any infection for individuals from the Copenhagen General Population Study with lymphopenia using age-adjusted percentiles of lymphocyte count.
Multivariable adjusted risk of any infection for individuals from the Copenhagen General Population Study with lymphopenia (lymphocyte count 9/l) compared to individuals with lymphocytes in the reference range (1.1–3.7 × 109/l). Multivariable adjustment includes all covariates listed in Table 1 except age, but with age as the underlying timescale. Individuals with lymphopenia had a lymphocyte count below the 2.5th percentile in each 1-year age band. aAccording to a likelihood ratio test.
Fig 7. Multivariable adjusted risk of any…
Fig 7. Multivariable adjusted risk of any infection for individuals from the Copenhagen General Population Study with lymphopenia stratified by follow-up time.
Multivariable adjusted risk of any infection for individuals from the Copenhagen General Population Study with lymphopenia (lymphocyte count 9/l) compared to individuals with lymphocyte count in the reference range (1.1–3.7 × 109/l). Multivariable adjustment includes all covariates listed in Table 1 except age, but with age as the underlying timescale. Follow-up time was segmented into 3 time intervals, and each individual was included in more than 1 time interval if total follow-up was above 2 years. When categorizing lymphocyte counts, lymphopenia was defined as a lymphocyte count below the 2.5th percentile, while the reference category was defined as a lymphocyte count between the 2.5th and 97.5th percentile. aAccording to a likelihood ratio test.
Fig 8. Algorithm.
Fig 8. Algorithm.
Absolute 2-year risks of hospitalization due to infection for different blood lymphocyte categories, stratified for age, sex, and smoking status. Nonsmokers were former smokers and never-smokers combined. Individuals diagnosed with immunodeficiency, hematologic disease, and/or autoimmune disease were not included when calculating absolute risks for the algorithm, since these individuals have a well-known high risk of infection. Numbers indicate absolute 2-year risk as rounded percentages.
Fig 9. Multivariable adjusted risk of infection-related…
Fig 9. Multivariable adjusted risk of infection-related death as a function of lymphocyte count for individuals from the Copenhagen General Population study.
The solid red line is the multivariable adjusted hazard ratio, and black dashed lines indicate the 95% confidence interval based on fitting of cubic splines to risk estimates obtained using Cox proportional hazards regression. Multivariable adjustment includes all covariates listed in Table 1 except age, but with age as the underlying timescale. The median lymphocyte value of 2.1 × 109/l was set as reference for the continuous model. When categorizing lymphocyte counts, lymphopenia was defined as a lymphocyte count below the 2.5th percentile, the reference category was defined as a lymphocyte count between the 2.5th and 97.5th percentile, and lymphocytosis was defined as a lymphocyte count above the 97.5th percentile.

References

    1. Tesfa D, Keisu M, Palmblad J. Idiosyncratic drug-induced agranulocytosis: possible mechanisms and management. Am J Hematol. 2009;84:428–34. 10.1002/ajh.21433
    1. Andersen CL, Tesfa D, Siersma VD, Sandholdt H, Hasselbalch H, Bjerrum OW, et al. Prevalence and clinical significance of neutropenia discovered in routine complete blood cell counts: a longitudinal study. J Intern Med. 2016;279:566–75. 10.1111/joim.12467
    1. Gibson C, Berliner N. How we evaluate and treat neutropenia in adults. Blood. 2014;124:1251–8. 10.1182/blood-2014-02-482612
    1. Boxer LA. How to approach neutropenia. Hematol Am Soc Hematol Educ Program. 2012;2012:174–82. 10.1182/asheducation-2012.1.174
    1. Stephan JL, Vlekova V, Deist F Le, Blanche S, Donadieu J, De Saint-Basile G, et al. Severe combined immunodeficiency: a retrospective single-center study of clinical presentation and outcome in 117 patients. J Pediatr. 1993;123:564–72. 10.1016/S0022-3476(05)80951-5
    1. Savino W. The thymus gland is a target in malnutrition. Eur J Clin Nutr. 2002;56:S46–9. 10.1038/sj.ejcn.1601485
    1. Zuluaga P, Sanvisens A, Teniente A, Fuster D, Tor J, Martinez-Caceres E, et al. Wide array of T-cell subpopulation alterations in patients with alcohol use disorders. Drug Alcohol Depend. 2016;162:124–9. 10.1016/j.drugalcdep.2016.02.046
    1. Matos LC, Batista P, Monteiro N, Ribeiro J, Cipriano MA, Henriques P, et al. Lymphocyte subsets in alcoholic liver disease. World J Hepatol. 2013;5:46–55. 10.4254/wjh.v5.i2.46
    1. Gergely P. Drug-induced lymphopenia: focus on CD4+ and CD8+ cells. Drug Saf. 1999;21:91–100. 10.2165/00002018-199921020-00003
    1. Ray-Coquard I, Cropet C, Van Glabbeke M, Sebban C, Le Cesne A, Judson I, et al. Lymphopenia as a prognostic factor for overall survival in advanced carcinomas, sarcomas, and lymphomas. Cancer Res. 2009;69:5383–91. 10.1158/0008-5472.CAN-08-3845
    1. Ownby HE, Roi LD, Isenberg RR, Brennan MJ. Peripheral lymphocyte and eosinophil counts as indicators of prognosis in primary breast cancer. Cancer. 1983;52:126–30.
    1. Fogar P, Sperti C, Basso D, Sanzari MC, Greco E, Davoli C, et al. Decreased total lymphocyte counts in pancreatic cancer: an index of adverse outcome. Pancreas. 2006;32:22–8.
    1. Greer JP, Foerster J, Rodgers GM, Paraskevas F, Gladder B, Arber DA, et al. Wintrobe’s clinical hematology. Volume 2 12th edition Philadelphia: Lippincott Williams & Wilkins; 2009.
    1. Merayo-Chalico J, Gómez-Martín D, Piñeirúa-Menéndez A, Santana-de Anda K, Alcocer-Varela J. Lymphopenia as risk factor for development of severe infections in patients with systemic lupus erythematosus: a case-control study. QJM. 2013;106:451–7. 10.1093/qjmed/hct046
    1. Ng WL, Chu CM, Wu AKL, Cheng VCC, Yuen KY. Lymphopenia at presentation is associated with increased risk of infections in patients with systemic lupus erythematosus. QJM. 2006;99:37–47. 10.1093/qjmed/hci155
    1. Ahmad DS, Esmadi M, Steinmann WC. Idiopathic CD4 lymphocytopenia: spectrum of opportunistic infections, malignancies, and autoimmune diseases. Avicenna J Med. 2013;3:37–47. 10.4103/2231-0770.114121
    1. Castelino DJ, McNair P, Kay TWH. Lymphocytopenia in a hospital population—what does it signify? Aust N Z J Med. 1997;27:170–4.
    1. Helby J, Nordestgaard BG, Benfield T, Bojesen SE. Shorter leukocyte telomere length is associated with higher risk of infections: a prospective study of 75,309 individuals from the general population. Haematologica. 2017;102:1457–65. 10.3324/haematol.2016.161943
    1. Nordestgaard BG, Palmer TM, Benn M, Zacho J, Tybjærg-Hansen A, Smith GD, et al. The effect of elevated body mass index on ischemic heart disease risk: causal estimates from a mendelian randomisation approach. PLoS Med. 2012; 9(5) :e1001212. 10.1371/journal.pmed.1001212
    1. Jørgensen AB, Frikke-Schmidt R, Nordestgaard BG, Tybjærg-Hansen A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med. 2014;371:32–41. 10.1056/NEJMoa1308027
    1. Pedersen CB, Gøtzsche H, Møller JØ, Mortensen PB. The Danish Civil Registration System. A cohort of eight million persons. Dan Med Bull. 2006;53:441–9. 10.1177/1403494810387965
    1. Sauce D, Larsen M, Fastenackels S, Roux A, Gorochov G, Katlama C, et al. Lymphopenia-driven homeostatic regulation of naive T cells in elderly and thymectomized young adults. J Immunol. 2012;189:5541–8. 10.4049/jimmunol.1201235
    1. Givi ME, Folkerts G, Wagenaar GTM, Redegeld FA, Mortaz E. Cigarette smoke differentially modulates dendritic cell maturation and function in time. Respir Res. 2015;16:131 10.1186/s12931-015-0291-6
    1. Apostolopoulos V, de Courten MPJ, Stojanovska L, Blatch GL, Tangalakis K, de Courten B. The complex immunological and inflammatory network of adipose tissue in obesity. Mol Nutr Food Res. 2016;60:43–57. 10.1002/mnfr.201500272
    1. Feldman C, Anderson R. Cigarette smoking and mechanisms of susceptibility to infections of the respiratory tract and other organ systems. J Infect. 2013;67:169–84. 10.1016/j.jinf.2013.05.004
    1. Campitelli MA, Rosella LC, Kwong JC. The association between obesity and outpatient visits for acute respiratory infections in Ontario, Canada. Int J Obese (Lond). 2014;38:113–9. 10.1038/ijo.2013.57
    1. Bueno V, Sant’Anna OA, Lord JM. Ageing and myeloid-derived suppressor cells: possible involvement in immunosenescence and age-related disease. Age (Dordr). 2014;36:9729 10.1007/s11357-014-9729-x
    1. Saroha S, Uzzo RG, Plimack ER, Ruth K, Al-Saleem T. Lymphopenia is an independent predictor of inferior outcome in clear cell renal carcinoma. J Urol. 2013;189:454–61. 10.1016/j.juro.2012.09.166
    1. Naushad H. Leukocyte count (WBC). Medscape; 2018 [cited 2018 Sep 20]. Available from: .
    1. Lynge E, Sandegaard JL, Rebolj M. The Danish National Patient Register. Scand J Public Health. 2011;39:30–3. 10.1177/1403494811401482
    1. Helweg-Larsen K. The Danish Register of Causes of Death. Scand J Public Health. 2011;39:26–9. 10.1177/1403494811399958
    1. Benfield T, Jensen JS, Nordestgaard BG. Influence of diabetes and hyperglycaemia on infectious disease hospitalisation and outcome. Diabetologia. 2007;50:549–54. 10.1007/s00125-006-0570-3
    1. Chang CJ, Chen LY, Liu LK, Lin MH, Peng LN, Chen LK. Lymphopenia and poor performance status as major predictors for infections among residents in long-term care facilities (LTCFs): a prospective cohort study. Arch Gerontol Geriatr. 2014;58:440–5. 10.1016/j.archger.2013.12.001
    1. Raben M, Walach N, Galili U, Schlesinger M. The effect of radiation therapy on lymphocyte subpopulations in cancer patients. Cancer. 1976;37:1417–21. 10.1002/cncr.23292
    1. Charlson ME, Pompei P, Ales KL, MacKenzie R. A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis. 1987;40:373–83.
    1. Quan H, Li B, Couris CM, Fushimi K, Graham P, Hider P, et al. Updating and validating the charlson comorbidity index and score for risk adjustment in hospital discharge abstracts using data from 6 countries. Am J Epidemiol. 2011;173:676–82. 10.1093/aje/kwq433
    1. Quan H, Sundararajan V, Halfon P, Fong A, Burnand B, Luthi J-C, et al. Coding algorithms for defining comorbidities in ICD-9-CM and ICD-10 administrative data. Med Care. 2005;43:1130–9.
    1. Eaton WW, Pedersen MG, Nielsen PR, Mortensen PB. Autoimmune diseases, bipolar disorder, and non-affective psychosis. Bipolar Disord. 2010;12:638–46. 10.1111/j.1399-5618.2010.00853.x
    1. Clarke R, Shipley M, Lewington S, Youngman L, Collins R, Marmot M, et al. Underestimation of risk associations due to regression dilution in long-term follow-up of prospective studies. Am J Epidemiol. 1999;150:341–53. 10.1093/oxfordjournals.aje.a010013
    1. Kotz S, Johnson N. Breakthroughs in statistics volume 1. Foundations and basic theory Springer Series in Statistics. New York: Springer; 1991.
    1. Andrès E, Maloisel F. Idiosyncratic drug-induced agranulocytosis or acute neutropenia. Curr Opin Hematol. 2008;15:15–21. 10.1097/MOH.0b013e3282f15fb9
    1. Hsieh MM, Everhart JE, Byrd-Holt DD, Tisdale JF, Rodgers GP. Prevalence of neutropenia in the U.S. population: age, sex, smoking status, and ethnic differences. Ann Intern Med. 2007;146:486–92.
    1. Palmblad J, Dufour C, Papadaki HA. How we diagnose neutropenia in the adult and elderly patient. Haematologica. 2014;99:1130–3. 10.3324/haematol.2014.110288
    1. Adrie C, Lugosi M, Sonneville R, Souweine B, Ruckly S, Cartier J-C, et al. Persistent lymphopenia is a risk factor for ICU-acquired infections and for death in ICU patients with sustained hypotension at admission. Ann Intensive Care. 2017;7:30 10.1186/s13613-017-0242-0
    1. Bain BJ. Ethnic and sex differences in the total and differential white cell count and platelet count. J Clin Pathol. 1996;49:664–6. 10.1136/jcp.49.8.664
    1. Tollerud DJ, Clark JW, Brown LM, Neuland CY, Pankiw-Trost LK, Blattner WA, et al. The influence of age, race, and gender on peripheral blood mononuclear-cell subsets in healthy nonsmokers. J Clin Immunol. 1989;9:214–22. 10.1007/BF00916817
    1. Qin L, Jing X, Qiu Z, Cao W, Jiao Y, Routy J-P, et al. Aging of immune system: immune signature from peripheral blood lymphocyte subsets in 1068 healthy adults. Aging (Albany NY). 2016;8:848–59. doi:
    1. Cicin-Sain L, Smyk-Pearson S, Currier N, Byrd L, Koudelka C, Robinson T, et al. Loss of naive T cells and repertoire constriction predict poor response to vaccination in old primates. J Immunol. 2010;184:6739–45. 10.4049/jimmunol.0904193
    1. McElhaney JE. Prevention of infectious diseases in older adults through immunization: the challenge of the senescent immune response. Expert Rev Vaccines. 2009;8:593–606. 10.1586/erv.09.12
    1. Yager EJ, Ahmed M, Lanzer K, Randall TD, Woodland DL, Blackman MA. Age-associated decline in T cell repertoire diversity leads to holes in the repertoire and impaired immunity to influenza virus. J Exp Med. 2008;205:711–23. 10.1084/jem.20071140
    1. Koch S, Larbi A, Ozcelik D, Solana R, Gouttefangeas C, Attig S, et al. Cytomegalovirus infection: a driving force in human T cell immunosenescence. Ann N Y Acad Sci. 2007;1114:23–35. 10.1196/annals.1396.043
    1. Linton PJ, Dorshkind K. Age-related changes in lymphocyte development and function. Nat Immunol. 2004;5:133–9. 10.1038/ni1033
    1. Calarota SA, Zelini P, De Silvestri A, Chiesa A, Comolli G, Sarchi E, et al. Kinetics of T-lymphocyte subsets and posttransplant opportunistic infections in heart and kidney transplant recipients. Transplantation. 2012;93:112–9. 10.1097/TP.0b013e318239e90c
    1. Baum PD, Young JJ, Schmidt D, Zhang Q, Hoh R, Busch M, et al. Blood T-cell receptor diversity decreases during the course of HIV infection, but the potential for a diverse repertoire persists. Blood. 2012;119:3469–78. 10.1182/blood-2011-11-395384
    1. Fülöp T, Gagné D, Goulet AC, Desgeorges S, Lacombe G, Arcand M, et al. Age-related impairment of p56(lck) and ZAP-70 activities in human T lymphocytes activated through the TcR/CD3 complex. Exp Gerontol. 1999;34:197–216. 10.1016/S0531-5565(98)00061-8
    1. Conroy RM, Pyörälä K, Fitzgerald AP, Sans S, Menotti A, De Backer G, et al. Estimation of ten-year risk of fatal cardiovascular disease in Europe: the SCORE project. Eur Heart J. 2003;24:987–1003. 10.1016/S0195-668X(03)00114-3

Source: PubMed

3
Iratkozz fel