Ratio of monocytes to lymphocytes in peripheral blood identifies adults at risk of incident tuberculosis among HIV-infected adults initiating antiretroviral therapy

Vivek Naranbhai, Adrian V S Hill, Salim S Abdool Karim, Kogieleum Naidoo, Quarraisha Abdool Karim, George M Warimwe, Helen McShane, Helen Fletcher, Vivek Naranbhai, Adrian V S Hill, Salim S Abdool Karim, Kogieleum Naidoo, Quarraisha Abdool Karim, George M Warimwe, Helen McShane, Helen Fletcher

Abstract

Background: Eight decades ago, the ratio of monocytes to lymphocytes (hereafter, the "ML ratio") was noted to affect outcomes of mycobacterial infection in rabbits. Recent transcriptomic studies support a role for relative proportions of myeloid and lymphoid transcripts in tuberculosis outcomes. The ML ratio in peripheral blood is known to be governed by hematopoietic stem cells with distinct biases.

Methods: The predictive value of the baseline ML ratio was modeled in 2 prospective cohorts of HIV-infected adults starting cART in South Africa (primary cohort, 1862 participants; replication cohort, 345 participants). Incident tuberculosis was diagnosed with clinical, radiographic, and microbiologic methods per contemporary guidelines. Kaplan-Meier survival analyses and Cox proportional hazards modeling were conducted.

Results: The incidence rate of tuberculosis differed significantly by baseline ML ratio: 32.61 (95% confidence interval [CI], 15.38-61.54), 16.36 (95% CI, 12.39-21.23), and 51.80 (95% CI, 23.10-101.71) per 1000 patient-years for ML ratios of less than the 5th percentile, between the 5th and 95th percentiles, and greater than the 95th percentile, respectively (P = .007). Neither monocyte counts nor lymphocyte counts alone were associated with tuberculosis. After adjustment for sex, World Health Organization human immunodeficiency virus disease stage, CD4(+) T-cell counts, and previous history of tuberculosis, hazards of disease were significantly higher for patients with ML ratios of less than the 5th percentile or greater than the 95th percentile (adjusted hazard ratio, 2.47; 95% CI, 1.39-4.40; P = .002).

Conclusions: The ML ratio may be a useful, readily available tool to stratify the risk of tuberculosis and suggests involvement of hematopoietic stem cell bias in tuberculosis pathogenesis.

Keywords: HIV; ML ratio; combination antiretroviral therapy; lymphocytes; monocytes; tuberculosis.

Figures

Figure 1.
Figure 1.
Kaplan-Meier estimates of probability of tuberculosis-free survival for individuals commencing combination antiretroviral therapy (cART), by category of baseline ratio of monocytes to lymphocytes (ML ratio; A). Bootstrapped hazard ratio (HR) estimates of tuberculosis across the ML ratio continuum (B). HRs are adjusted for age, World Health Organization human immunodeficiency virus infection and disease stage, and past history of tuberculosis. Each dot denotes 1 estimate, with the locally weighted scatterplot smoothing curve overplotted.
Figure 2.
Figure 2.
Ratios of monocytes to lymphocytes (ML ratios), monocyte counts, and lymphocyte counts among healthy human immunodeficiency virus–uninfected woman. A, The correlation between the ML ratio at enrollment and 3, 12, and 24 months after enrollment among 790 participants. Results of Spearman rank correlation are shown as rho and P values in the top right of each correlation plot. B, The overall stability of ML ratios in healthy individuals, shown as box plots at each time point. The dotted horizontal lines denote the overall population 5th and 95th percentiles. Boxes show the 25th, 50th (median), and 75th percentiles, whiskers denote the 25th percentile – [1.5 × interquartile range] and the 75th percentile + [1.5 × interquartile range], and outliers are plotted as dots. C, ML ratios may be abnormal even if the monocyte or lymphocyte counts are normal. Monocyte and lymphocyte counts are plotted on the x-axes and y-axes, respectively. Colors of data points denote the corresponding category of the ML ratio (5th–95th percentiles, >95th percentile, and < 5th percentile). Dotted horizontal lines denote the 5th and 95th percentiles for lymphocyte counts. Dotted vertical lines denote the 5th and 95th percentiles for monocyte counts. Abbreviation: NS, not significant.
Figure 3.
Figure 3.
The impact of human immunodeficiency virus (HIV) acquisition and disease progression on ratios of monocytes to lymphocytes (ML ratios). A, The ML ratio is stable before infection among 160 women who acquired HIV but is significantly increased by HIV acquisition. Boxes show the 25th, 50th (median), and 75th percentiles, whiskers denote the 25th percentile – [1.5 × interquartile range] and the 75th percentile + [1.5 × interquartile range], and outliers are plotted as dots. B, The distribution of ML ratios is broadened by HIV acquisition. C, ML ratios over the course of HIV acquisition are variable but increase overall. Data are shown as a scatterplot showing all ML measures over time among HIV acquirers. The black line is a smoothed curve computed by locally weighted scatterplot smoothing. Horizontal lines denote the 5th and 95th percentile for ML ratios in HIV-uninfected healthy individuals in red and blue, respectively. D, ML ratios over the course of HIV infection increase significantly with disease progression. Boxes show the 25th, 50th (median), and 75th percentiles, whiskers denote the 25th percentile – [1.5 × interquartile range] and the 75th percentile + [1.5 × interquartile range], and outliers are plotted as dots. Horizontal lines denote the 5th and 95th percentile for ML ratios in HIV-uninfected healthy individuals in red and blue, respectively.
Figure 4.
Figure 4.
The impact of combination antiretroviral therapy (cART) on the ratio of monocytes to lymphocytes (ML ratio). A, The ML ratio is reduced significantly over the first 2 years of cART and stabilizes within the normal range for human immunodeficiency virus (HIV)–uninfected individuals until up to 7 years after cART commencement. Box plots show the ML ratio before and during 7 years of cART. Boxes show the 25th, 50th (median), and 75th percentiles, whiskers denote the 25th percentile – [1.5 × interquartile range] and the 75th percentile + [1.5 × interquartile range], and outliers are plotted as dots. Horizontal lines denote the 5th and 95th percentile for ML ratios in HIV-uninfected healthy individuals in red and blue, respectively. B, The distribution of ML ratios becomes narrower over time.

References

    1. Cohen T, Murray M, Wallengren K, Alvarez GG, Samuel EY, Wilson D. The prevalence and drug sensitivity of tuberculosis among patients dying in hospital in KwaZulu-Natal, South Africa: a postmortem study. PLoS Med. 2010;7:e1000296.
    1. Cox JA, Lukande RL, Lucas S, Nelson AM, Van Marck E, Colebunders R. Autopsy causes of death in HIV-positive individuals in sub-Saharan Africa and correlation with clinical diagnoses. AIDS Rev. 2010;12:183–94.
    1. Rana FS, Hawken MP, Mwachari C, et al. Autopsy study of HIV-1-positive and HIV-1-negative adult medical patients in Nairobi, Kenya. J Acquir Immune Defic Syndr. 2000;24:23–9.
    1. Walzl G, Ronacher K, Hanekom W, Scriba TJ, Zumla A. Immunological biomarkers of tuberculosis. Nat Rev Immunol. 2011;11:343–54.
    1. Lawn SD, Zumla AI. Tuberculosis. Lancet. 2011;378:57–72.
    1. Rangaka MX, Wilkinson KA, Glynn JR, et al. Predictive value of interferon-gamma release assays for incident active tuberculosis: a systematic review and meta-analysis. Lancet Infect Dis. 2012;12:45–55.
    1. Samandari T, Agizew TB, Nyirenda S, et al. 6-month versus 36-month isoniazid preventive treatment for tuberculosis in adults with HIV infection in Botswana: a randomised, double-blind, placebo-controlled trial. Lancet. 2011;377:1588–98.
    1. Lawn SD, Wood R, De Cock KM, Kranzer K, Lewis JJ, Churchyard GJ. Antiretrovirals and isoniazid preventive therapy in the prevention of HIV-associated tuberculosis in settings with limited health-care resources. Lancet Infect Dis. 2010;10:489–98.
    1. Madhi SA, Nachman S, Violari A, et al. Primary isoniazid prophylaxis against tuberculosis in HIV-exposed children. N Engl J Med. 2011;365:21–31.
    1. Berry MP, Graham CM, McNab FW, et al. An interferon-inducible neutrophil-driven blood transcriptional signature in human tuberculosis. Nature. 2010;466:973–7.
    1. Maertzdorf J, Repsilber D, Parida SK, et al. Human gene expression profiles of susceptibility and resistance in tuberculosis. Genes Immun. 2011;12:15–22.
    1. Cunningham RS, Sabin FR, Sugiyama S, Kindwall JA. The role of the monocyte in tuberculosis. Bull Johns Hopkins Hosp. 1925;XXXVII:231–80.
    1. Sabin FR, Doan CA, Cunningham RS. Studies of the blood in experimental tuberculosis: the monocyte-lymphocyte ratio; the anemia-leucopenia phase. Trans 22nd Annu Meeting Natl Tuberc Assoc; 1926. pp. 252–6.
    1. Rogers PM. A study of the blood monocytes in children with tuberculosis. N Engl J Med. 1928;198:740–49.
    1. Doan CA, Sabin FR. The relation of the tubercle and the monocyte:lymphocte ratio to resistance and susceptibility in tuberculosis. J Exp Med. 1930;52:113–52.
    1. Naranbhai V, Abdool Karim Q, Naidoo K, Yende Zuma N, Abdool Karim SS. Task-shifting for antiretroviral treatment is not only effective but also sustainable and improves over time. Lancet. 2012;380:1907–8.
    1. Abdool Karim Q, Abdool Karim SS, Frohlich JA, et al. Effectiveness and safety of tenofovir gel, an antiretroviral microbicide, for the prevention of HIV infection in women. Science. 2010;329:1168–74.
    1. Abdool Karim SS, Naidoo K, Grobler A, et al. Timing of initiation of antiretroviral drugs during tuberculosis therapy. N Engl J Med. 2010;362:697–706.
    1. van Loggerenberg F, Mlisana K, Williamson C, et al. Establishing a cohort at high risk of HIV infection in South Africa: challenges and experiences of the CAPRISA 002 acute infection study. PLoS One. 2008;3:e1954.
    1. Republic of South Africa Department of Health of South Africa, Pretoria, South Africa. South African National Antiretroviral Treatment Guidelines. 2004.
    1. Republic of South Africa Department of Health of South Africa, Pretoria, South Africa. South African National Tuberculosis Management Guidelines. 2009.
    1. Republic of South Africa Department of Health of South Africa, Pretoria, South Africa. South African National Antiretroviral Treatment Guidelines. 2010.
    1. Abdool Karim SS, Naidoo K, Grobler A, et al. Integration of antiretroviral therapy with tuberculosis treatment. N Engl J Med. 2011;365:1492–501.
    1. Lawn SD, Badri M, Wood R. Tuberculosis among HIV-infected patients receiving HAART: long term incidence and risk factors in a South African cohort. AIDS. 2005;19:2109–16.
    1. Dembele M, Saleri N, Carvalho AC, et al. Incidence of tuberculosis after HAART initiation in a cohort of HIV-positive patients in Burkina Faso. Int J Tuberc Lung Dis. 2010;14:318–23.
    1. Seyler C, Toure S, Messou E, Bonard D, Gabillard D, Anglaret X. Risk factors for active tuberculosis after antiretroviral treatment initiation in Abidjan. Am J Respir Crit Care Med. 2005;172:123–7.
    1. Carpenter E, Fray L, Gormley E. Cellular responses and Mycobacterium bovis BCG growth inhibition by bovine lymphocytes. Immunol Cell Biol. 1997;75:554–60.
    1. Denis M, Wedlock DN, Buddle BM. Ability of T cell subsets and their soluble mediators to modulate the replication of Mycobacterium bovis in bovine macrophages. Cell Immunol. 2004;232:1–8.
    1. Tobin DM, Roca FJ, Oh SF, et al. Host genotype-specific therapies can optimize the inflammatory response to mycobacterial infections. Cell. 2012;148:434–46.
    1. Casadevall A, Pirofski LA. The damage-response framework of microbial pathogenesis. Nat Rev Microbiol. 2003;1:17–24.
    1. Merekoulias G, Alexopoulos EC, Belezos T, Panagiotopoulou E, Jelastopulu DM. Lymphocyte to monocyte ratio as a screening tool for influenza. PLoS Curr. 2010;2:RRN1154.
    1. Muller-Sieburg CE, Cho RH, Thoman M, Adkins B, Sieburg HB. Deterministic regulation of hematopoietic stem cell self-renewal and differentiation. Blood. 2002;100:1302–9.
    1. Pang WW, Price EA, Sahoo D, et al. Human bone marrow hematopoietic stem cells are increased in frequency and myeloid-biased with age. Proc Natl Acad Sci U S A. 2011;108:20012–7.
    1. Schroeder T. Hematopoietic stem cell heterogeneity: subtypes, not unpredictable behavior. Cell Stem Cell. 2010;6:203–7.
    1. Muller-Sieburg CE, Sieburg HB, Bernitz JM, Cattarossi G. Stem cell heterogeneity: implications for aging and regenerative medicine. Blood. 2012;119:3900–7.
    1. Baldridge MT, King KY, Boles NC, Weksberg DC, Goodell MA. Quiescent haematopoietic stem cells are activated by IFN-gamma in response to chronic infection. Nature. 2010;465:793–7.
    1. Das B, Kashino SS, Pulu I, et al. CD271+ bone marrow mesenchymal stem cells may provide a niche for dormant Mycobacterium tuberculosis. Sci Transl Med. 2013;5 170ra13-ra13.
    1. Gideon HP, Flynn JL. Latent tuberculosis: what the host “sees”? Immunol Res. 2011;50:202–12.
    1. Kato K. Effect of tuberculin test on monocyte-lymphocyte ratio in children. J Pediatr. 1935;7:238–44.
    1. Warimwe GM, Murungi LM, Kamuyu G, et al. The ratio of monocytes to lymphocytes in peripheral blood correlates with increased susceptibility to clinical malaria in Kenyan children. PLoS One. 2013;8:e57320.

Source: PubMed

3
Subscribe