Abstract
Objective
Since immune dysfunction is thought to underlie the development of Non-Hodgkin Lymphoma (NHL), obesity and chronic inflammation may be involved in its etiology. We examined the association of pre-diagnostic inflammatory markers and adipokines with NHL risk.
Methods
We conducted a nested case-control analysis (272 cases and 541 matched controls) within the Multiethnic Cohort. Luminex technology was used to measure a 10-plex panel of cytokines, ELISA assays for adipokines, and an autoanalyzer for C-reactive protein (CRP). Odds ratios (ORs) and 95% confidence intervals (CIs) for tertiles of analytes were estimated by conditional logistic regression.
Results
After a median time of 2.7 years from phlebotomy to diagnosis, interleukin (IL)-10 was significantly related to NHL risk (ORT3 vs T1=3.07; 95%CI: 2.02–4.66; ptrend <0.001). TNF-α and IL-8 showed borderline elevated risks, while IFN-γ, IL-1β, IL-2, IL-4, IL-5, IL-6, and CRP were not associated with NHL. Leptin but not adiponectin was related to NHL risk (ORT3 vs T1=0.48, 95%CI: 0.30–0.76; ptrend<0.001). Adjustment for body mass index did not substantially affect the risk estimates. Stratification by subtype indicated significant associations with IL-10 and leptin for follicular but not for diffuse large B-cell lymphoma. Excluding cases diagnosed <1 year after phlebotomy attenuated all associations.
Conclusions
IL-10 was the only cytokine and leptin the only adipokine associated with NHL, but, due to the short follow-up time, pre-clinical effects cannot be excluded.
Impact
Although markers of inflammation and adiposity may provide new insights into the etiology of NHL, they need to be assessed many years before clinical diagnosis.
Keywords: Non-Hodgkin Lymphoma, inflammatory markers, adiponectin, leptin, nested case-control study, prospective cohort, ethnicity
Introduction
Non-Hodgkin lymphoma (NHL) is an etiologically, clinically, and histologically heterogeneous group of malignant diseases of the lymphocytes (1). Although the etiology of NHL remains unclear, subclinical immune dysfunction is the most consistent risk factor (2–5) and dysregulation of cytokines may mediate disease progression (6). Cytokines regulate lymphocyte development and differentiation; imbalance in the expression of cytokines from T helper 1 (Th1) and Th2 lymphocytes may play an important role in the pathogenesis of NHL (7, 8). Specifically, Th1 cytokines, e.g., tumor necrosis factor (TNF)-α, interleukin (IL)-2, interferon (IFN)-γ, may promote cell-mediated immune response, while Th2 cytokines, e.g., Il-4, IL-5, IL-6, IL1-β, and IL-13, may be involved in the humoral immune response and favor B-cell activation (9). Independent of pathways required for Th1 and Th2 cell development, regulatory T cells (Treg) and IL-17-secreting Th cells (Th17) also play a critical role in autoimmunity (10, 11). As Th17 and Treg cells constitute two opposing immune responses that are critically involved in the modulation of inflammation, a lack of Th17 cells present in the tumor microenvironment of NHL and an imbalance between the two cell types may contribute to NHL etiology (12). Genetic polymorphisms in several cytokine genes, i.e., TNF, IL-4, IL-5, IL-6, IL-10, have been associated with susceptibility to NHL (13–19). However, only a few prospective studies have evaluated pre-diagnostic cytokines and subsequent NHL risk in immunocompetent populations (20–22). These studies observed positive associations for IL-10 (20) and TNF-α (20, 21) and inverse associations for IL-2 (22), IL-5 (21, 22), and IFN-γ (22).
Obesity results in a pathological state of chronic low-level inflammation and altered immune responses that may influence B- and T-lymphocyte function (23) and, thus, the development of NHL. However, the results for obesity and NHL in epidemiologic studies are inconsistent. In one meta-analysis with 10 cohorts and 6 case-control studies, overweight individuals had a 7% greater and obese individuals a 20% greater risk of NHL than normal weight individuals (24), but a pooled analysis of 10,000 cases and 16,000 controls across 18 case-control studies detected only a small excess risk for diffuse large B-cell lymphoma (DLBCL) (25). A recent meta-analysis of 16 prospective studies (26) reported a 7% higher risk for each 5 kg/m2 increase in BMI but only for DLBLC and not for other subtypes.
Adipocyte-derived adipokines may act as mediators in the NHL pathogenesis. Leptin participates in the inflammation response and enhances B-cell survival (27, 28). Adiponectin attenuates the production of pro-inflammatory cytokines, e.g., TNF-α in human macrophages (29), and increases immunosuppressive cytokines, such as IL-10, in human leukocytes (30). TNF-α may also have an anti-apoptotic effect on B-cells by promoting the activation of transcription factor nuclear factor-κB (31). Genetic polymorphisms in the TNF gene modified the association between body mass index (BMI) and risk for follicular lymphoma (FL) in one study (14), whereas only the risk for DLBCL and not for FL was elevated in obese individuals with specific genotypes of TNF-α or IL-10 supporting the hypothesis that an obesity-related chronic inflammatory response may play a role in the etiology of NHL and DLBCL (32). This case-control study nested within the Multiethnic Cohort (MEC) study explored the relation of pre-diagnostic serum levels of Th1/Th2 cytokines, leptin, adiponectin, and CRP with NHL risk. We hypothesized an elevated NHL risk with Th2, B-cell stimulating, and proinflammatory cytokines (IL-1β, IL-4, IL-5, IL-6, IL-10, TNF-α) and the chemokine IL-8, as well as with leptin, CRP, and protective effects for Th1 cytokines (IFN-γ, IL-2) and adiponectin.
Materials and Methods
Study population
The MEC is a longitudinal study designed to investigate the association of dietary, lifestyle, and genetic factors with the incidence of cancer. The cohort was assembled in Hawaii and Los Angeles in 1993–1996; details on recruitment and baseline information have been reported previously (33). Briefly, subjects from 5 ethnic groups (African American, Native Hawaiian, Japanese American, Latino, and white) were identified primarily through drivers’ license files supplemented with other sources and recruited by mailing a self-administered, 26-page questionnaire on diet, anthropometric measures, medical history, family history of cancer, and lifestyle. A total of 215,251 men and women aged 45 to 75 years were included at baseline and formed a group broadly representative of the general population as verified by a comparison of the cohort’s distributions across educational levels and marital status with corresponding census data (33). The study protocol was approved by the Institutional Review Boards of the University of Hawaii and the University of Southern California.
Specimen collection
The prospective MEC biospecimen subcohort was predominately established between 2001 and 2006 by asking surviving cohort members to provide specimens of blood and urine (34). In total, 67,594 cohort members contributed to the biorepository from which the cases and the controls were selected. When comparing the characteristics of repository are broadly representative of all cohort members (Table 1). The median processing times for blood samples was <6 hours and similar for cases and controls.
Table 1.
Characteristics | Casesa | Controlsa | p-valueb | MECc |
---|---|---|---|---|
N | 272 | 541 | 203,621 | |
Age at blood draw, yd | 70.0 ± 7.4 | 70.0 ± 7.5 | ||
Fasting prior to blood draw, hrs d | 12.0 ± 4.0 | 12.3 ± 4.1 | ||
Maled | 157 (58) | 313 (58) | 91,905 (45) | |
Ethnicityd | ||||
White | 74 (27) | 148 (27) | 49,604 (24) | |
Japanese American | 72 (27) | 143 (27) | 57,011 (28) | |
Latino | 62 (23) | 123 (23) | 47,516 (24) | |
African American | 47 (17) | 94 (17) | 34,971 (17) | |
Native Hawaiian | 17 (6) | 33 (6) | 14,519 (7) | |
BMI, kg/m2 | 27.0 ± 5.4 | 26.4 ± 4.4 | 0.11 | 26.1±5.1 |
Overweight (25.0–29.9 kg/m2) | 106 (39) | 219 (40) | 72,739 (36) | |
Obese (≥30.0 kg/m2) | 57 (21) | 96 (18) | 34,515 (17) | |
Secondary or higher education | 182 (67) | 345 (64) | 0.39 | 110, 471 (55) |
Alcohol intake ≥1 drink/month | 137 (50) | 284 (53) | 0.57 | 114,129 (56) |
Ever smoker | 164 (60) | 291 (54) | 0.08 | 112,185 (55) |
Pack-years of cigarette smoking | 10.4 ±15.1 | 9.8 ± 15.2 | 0.55 | 10.1±15.0 |
Diabetes, self-reported | 36 (13) | 22 (4) | <0.001 | 24,342 (12) |
Circulating inflammatory markers | ||||
Adiponectin, μg/mL | 7.4 (4.4–13.7) | 7.8 (4.7–13.0) | 0.71 | |
Leptin, ng/mL | 8.5 (3.8–17.1) | 10.6 (5.2–21.8) | <0.01 | |
IFN-γ, pg/mL | 0.7 (0.3–1.2) | 0.6 (0.4–1.0) | 0.97 | |
IL-1β, pg/mL | 1.1 (0.5–2.5) | 1.0 (0.5–2.1) | 0.38 | |
IL-2, pg/mL | 1.3 (0.7–2.5) | 1.3 (0.7–2.3) | 0.39 | |
IL-4, pg/mL | 2.3 (1.2–4.1) | 2.3 (1.1–4.2) | 0.62 | |
IL-5, pg/mL | 0.6 (0.3–1.2) | 0.6 (0.3–1.1) | 0.41 | |
IL-6, pg/mL | 1.9 (0.9–4.1) | 1.6 (0.9–3.42 | 0.13 | |
IL-8, pg/mL | 10.5 (5.9–20.3) | 9.2 (5.3–17.9) | 0.10 | |
IL-10, pg/mL | 1.4 (0.6–3.0) | 0.9 (0.5–1.7) | <0.001 | |
TNF-α, pg/mL | 2.6 (1.4–6.5) | 2.6 (1.2–5.9) | 0.41 | |
CRP, mg/L | 1.9 (0.6–4.7) | 1.8 (0.7–4.3) | 0.61 | |
Summary measuree | −0.2 (−0.6–0.3) | −0.6 (−0.2–0.3) | 0.49 |
Abbreviations: BMI, body mass index; IFN, Interferon; IL, Interleukin; TNF, Tumor neurosis factor; CRP, C-reactive protein.
Values are mean ± SD, n (%) or median (25th–75th percentile). Data were missing for fasting hours (8), education (4), alcohol (17), leptin (1), CRP (3) among NHL cases and controls and for BMI (2,853) and secondary or higher education (2,437) in MEC.
p-values from χ2 test, Students t-test, or Wilcoxon rank sum test.
Comparison based on entire MEC data set excluding “Other” ethnicity (N=203,621)
Matching criteria.
Composite score from principal components analysis of log-transformed IL-1β, IL-4, IL-5, IL-6, IL-8, and TNF-α.
Selection of cases and controls
Incident cancer cases within the MEC were identified by routine linkages to the Hawaii Tumor Registry, the Los Angeles County Cancer Surveillance Program, and the State of California Cancer Registry, which are part of the National Cancer Institute Surveillance, Epidemiology, and End Results (SEER) program. Only cases with a date of blood draw before diagnosis were included in the present study. NHL cases were categorized into common subtypes using the classification of lymphoid neoplasms recommended for epidemiologic research based on the International Classification of Diseases for Oncology, Third Edition coding (1). The common subtypes included DLBCL, small lymphocytic lymphoma or chronic lymphocytic leukemia (SLL/CLL), follicular lymphoma (FL), T-cell lymphoma (all types), and all other NHL subtypes. Controls were identified from the biospecimen subcohort who were alive and free of NHL at the age of the case’s diagnosis and matched on sex, birth date (within ±1 year), ethnicity (white, Japanese American, Latino, African American, or Native Hawaiian), location (California or Hawaii), date of blood draw (within ±1 year), time of day of blood draw, and fasting hours prior to blood draw (0–<6, 6–<8, 8–<10, or ≥10). The last two matching criteria were needed for analytes other than inflammatory markers. Through linkages with the state death certificate files in California and Hawaii and the National Death Index, we confirmed that the controls were alive at the date of diagnosis of their matched cases.
Lab Assays
Although 275 cases were selected for this analysis, 3 cases had missing serum values and another 3 cases had only 1 control value available resulting in a sample size of 272 cases and 541 controls. All plasma specimens from cases and controls were exposed to the same number of freeze-thaw cycles. The Luminex panel included 8 cytokines (IFN-γ, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-10, and TNF-α) and one chemokine (IL-8), which were measured using a modification of an Invitrogen (Carlsbad, CA) magnetic high sensitivity 10-plex assay kit (LHC0001) and Luminex 200 plate reader. We used 25 μL of serum and 75 μL of diluent buffer per well and decreased the quantity of beads by 25% from manufacturer directions in order to increase sensitivity and reduce clogging of the Luminex plate reader. Each plate held duplicate samples for its assigned subjects. A standard curve was generated using 25 μL per well for each serial dilution of the standard mix provided by the manufacturer. The buffer blank provided by the manufacturer was found to be unacceptable because many samples had lower fluorescence than the blank. Therefore, the lowest fluorescing serum value in each batch was used as a serum blank and subtracted from all samples. Since the fluorescence of many plasma samples was close to 0, using the lowest plasma sample in each batch gave a more reasonable zero value to obtain a relative comparison and non-negative values without significant deviation between batches. Only for IL-8 and Il-10, the buffer blank gave a low value and was used in the calculation as a blank. Values were calculated manually from the fluorescent values reported by the Luminex reader using a standard curve for each analyte plotted on a linear scale and adjusted for % recovery of each analyte determined from spiked serum samples. Extrapolation between 0 and the lower limit of detection (LLOD) was required for <10% of all samples; the rate was higher only for TNF-α and IFN-γ. From previous work with the same assay at our center, we had determined that the extrapolated values gave higher intraclass correlation coefficients than other options, i.e., setting all to 0, setting all values between 0 and LLOD to 1/2 of LLOD, and imputation of new values between 0 and LLOD using the observed distribution ignoring correlation structure between duplicates. Based on these results, we decided that using the extrapolated results between 0 and the LLODs gave more robust results than advertised by the manufacturer.
Leptin and adiponectin were measured utilizing human Leptin (Catalogue #DLP00) and Adiponectin (Catalogue #DRP300) immunoassay kits purchased from R&D Systems, (Minneapolis, MN) according to the manufacturer’s directions with detection on a Molecular Devices Versamax tunable microplate reader with Softmax Pro analysis software (Molecular Devices Corp., Sunnyvale, CA) using a 5-parameter analysis of the resulting standard curve. CRP was assessed using a Cobas MiraPlus clinical chemistry analyzer.
The LLODs were as follows: 0.1 pg/mL for IL-1β; 0.2 pg/mL for IL-2, IL-4, IL-5, IL-6, and IL-10; 0.5 pg/mL for IL-8; 0.6 pg/mL for IFN-γ; 1.0 pg/mL for TNF-α; 1.6 ng/mL for leptin; 0.4 μg/mL for adiponectin; and 0.1 mg/L for CRP. Replicate samples of pooled serum were included in each analysis batch for each analyte for quality control. Based on 27 duplicate and 9 triplet samples, the respective within- and between-batch coefficients of variation based on log-transformed marker values were as follows: adiponectin (2.5 and 11.2%); leptin (3.4 and 4.6%); IFN-γ (15.0 and 21.6%); IL-1β (20.9 and 40.7%); IL-2 (10.3 and 16.2%); IL-4 (9.2 and 10.0%); IL-5 (7.1 and 31.1%); IL-6 (8.9 and 18.9%); IL-8 (3.8 and 10.9%); IL-10 (7.8 and 32.1%); TNF-α (10.0 and 43.2%); and CRP (3.5 and 3.2%).
Statistical analysis
SAS version 9.2 (SAS Institute, Inc., Cary, NC) was used to perform all statistical analyses, with a two-sided p value of <0.05 considered statistically significant. Cases and controls were compared using the χ2 test for categorical variables and the t-test or Wilcoxon rank sum test for continuous variables. Spearman correlation coefficients were used to determine the relation between BMI and analytes. Principal components analysis was used to address the correlation among the cytokines and to create a composite score. The number of components retained for orthogonal rotation was based primarily on functional characteristics of cytokines, e.g., B-cell simulating, pro-inflammatory, with the requirement of an eigenvalue >1.0 and explained variance >60% (35). A factor was constructed as a linear composite of the cytokines with meaningful loading scores, e.g., ≥0.30, and used to calculate a score for each subject.
Using conditional logistic regression, odds ratios (OR) and 95% confidence intervals (95% CI) were estimated for tertiles of circulating markers and NHL risk. The tertiles were based on the exposure distribution of both cases and controls to ensure a sufficient number of cases and controls within strata. Linear trends were tested by modeling log-transformed continuous variables. Potential confounders, such as years of education, alcohol consumption, cigarette smoking, physical activity, history of blood transfusion, history of asthma, antihistamine, aspirin or acetaminophen use, and dietary intake of fiber, fruit, or vegetables, were examined, but were not included in the final models, as they were not found alone, or in combination, to change the risk estimates by more than 10% (36). For analytes associated with BMI, we repeated all models with and without adjustment for BMI. Sensitivity analyses to control for early disease effects were performed by excluding cases diagnosed <1 year after the blood draw. Heterogeneity of the risk estimates by ethnicity, sex, BMI (<25 vs. ≥ 25 kg/m2), and years between blood draw and NHL diagnosis (<2.7 vs. ≥ 2.7 years) was tested by a Wald test of cross-product terms. We also evaluated heterogeneity by common NHL subtype (DLBCL, SLL/CLL, and FL) by a Wald test of the parameter estimates obtained from unconditional polytomous logistic regression accounting for the matching factors and performed stratified analyses for DLBCL and FL.
Results
The incident NHL cases were primarily of B-cell origin, with 79 (29%) DLBCL, 51 (19%) SLL/CLL, 49 (18%) FL, 15 (6%) T-cell lymphoma, and 78 (28%) other NHL subtypes. Cases were aged 70.0±7.4 years at blood draw (Table 1), with a median of 2.7 (Interquartile Range 1.4–4.4; range 0.01–11.5) years between blood draw and diagnosis. Whites (27%) and Japanese Americans (27%) comprised the largest ethnic groups, followed by Latinos (23%), African Americans (17%), and Native Hawaiians (6%). Educational level, BMI, and alcohol consumption did not differ between cases and controls. Cases were more likely to be ever smokers (p=0.08) and to have a self-reported history of diabetes (p<0.001) compared to controls (Table 1). They also had statistically significantly lower pre-diagnostic serum levels of leptin (p<0.01) and higher levels of IL-10 (p<0.001) and marginally higher levels of IL-8 (p=0.10) than controls, whereas other markers did not differ by case status. Among controls, BMI was positively correlated with leptin (r=0.45, p<0.0001), CRP (r=0.29, p<0.0001), and IL-6 (r=0.08, p=0.05) and negatively correlated with adiponectin (r=−0.20, p<0.0001), IL-1β (r=−0.09, p=0.03), and TNF-α (r=−0.09, p=0.03).
Only pre-diagnostic serum leptin and IL-10 levels were significantly associated with NHL risk (Table 2). When comparing the highest vs. the lowest tertiles of leptin, NHL risk was reduced by 52% (ORT3 vs. T1=0.48 [0.30–0.76]; ptrend<0.001), whereas a 3-fold elevation in NHL risk was observed for IL-10 (ORT3 vs. T1=3.07 [2.02–4.66]; ptrend<0.001). Excluding cases diagnosed <1 year after the blood draw attenuated the risk estimates for leptin (ORT3 vs. T1=0.61 [0.37–1.02]; ptrend=0.05) and IL-10 (ORT3 vs. T1=2.20 [1.38–3.51]; ptrend=0.06). The overall associations with TNF-α (Ptrend= 0.05) and IL-8 (Ptrend= 0.08) were borderline significant, and none of the remaining markers or the summary measure were associated with NHL risk.
Table 2.
Analyte | T1 | T2 | T3 | ptrend b |
---|---|---|---|---|
Adiponectin, μg/mL | <5.6 | 5.6–<11.0 | ≥11.0 | |
Cases/controls | 98/177 | 84/184 | 90/180 | |
OR (95% CI) all | 1.00 | 0.81 (0.55, 1.17) | 0.87 (0.58, 1.30) | 0.67 |
OR (95% CI) >1 y onlyc | 1.00 | 0.78 (0.50, 1.19) | 0.87 (0.55, 1.36) | 0.63 |
Leptin, ng/ml | <6.4 | 6.4–<14.9 | ≥14.9 | |
Cases/controls | 105/166 | 91/182 | 76/192 | |
OR (95% CI) all | 1.00 | 0.72 (0.49, 1.05) | 0.48 (0.30, 0.76) | <0.001 |
OR (95% CI) >1 y onlyc | 1.00 | 0.73 (0.48, 1.13) | 0.61 (0.37, 1.02) | 0.05 |
IFN-γ, pg/mL | <0.5 | 0.5–<0.9 | ≥0.9 | |
Cases/controls | 93/172 | 80/195 | 99/174 | |
OR (95% CI) all | 1.00 | 0.75 (0.51, 1.10) | 1.05 (0.72, 1.55) | 0.61 |
OR (95% CI) >1 y onlyc | 1.00 | 0.69 (0.44, 1.06) | 1.02 (0.66, 1.57) | 0.82 |
IL-1β, pg/mL | <0.6 | 0.6–<1.6 | ≥1.6 | |
Cases/controls | 93/176 | 83/189 | 96/176 | |
OR (95% CI) all | 1.00 | 0.81 (0.54, 1.22) | 1.04 (0.69, 1.56) | 0.09 |
OR (95% CI) >1 y onlyc | 1.00 | 1.00 (0.64, 1.56) | 1.00 (0.63, 1.60) | 0.52 |
IL-2, pg/mL | <0.9 | 0.9–<1.9 | ≥1.9 | |
Cases/controls | 86/182 | 91/183 | 95/176 | |
OR (95% CI) all | 1.00 | 1.07 (0.72, 1.58) | 1.16 (0.79, 1.72) | 0.14 |
OR (95% CI) >1 y onlyc | 1.00 | 1.08 (0.69, 1.69) | 1.10 (0.70, 1.73) | 0.51 |
IL-4, pg/mL | <1.4 | 1.4–<3.4 | ≥3.4 | |
Cases/controls | 87/182 | 97/177 | 88/182 | |
OR (95% CI) all | 1.00 | 1.14 (0.79, 1.65) | 1.00 (0.65, 1.55) | 0.25 |
OR (95% CI) >1 y onlyc | 1.00 | 1.26 (0.83, 1.92) | 1.01 (0.62, 1.65) | 0.53 |
IL-5, pg/mL | <0.4 | 0.4–<0.9 | ≥0.9 | |
Cases/controls | 87/181 | 87/185 | 98/175 | |
OR (95% CI) all | 1.00 | 0.96 (0.60, 1.55) | 1.20 (0.71, 2.02) | 0.14 |
OR (95% CI) >1 y onlyc | 1.00 | 1.05 (0.70, 1.58) | 1.32 (0.84, 2.07) | 0.47 |
IL-6, pg/mL | <1.1 | 1.1–<2.7 | ≥2.7 | |
Cases/controls | 85/187 | 86/189 | 101/165 | |
OR (95% CI) all | 1.00 | 1.02 (0.71, 1.48) | 1.39 (0.95, 2.02) | 0.17 |
OR (95% CI) >1 y onlyc | 1.00 | 0.93 (0.62, 1.41) | 1.11 (0.72, 1.70) | 0.97 |
IL-8, pg/mL | <6.7 | 6.7–<14.6 | ≥14.6 | |
Cases/controls | 79/188 | 94/179 | 99/174 | |
OR (95% CI) all | 1.00 | 1.30 (0.88, 1.91) | 1.45 (0.97, 2.16) | 0.08 |
OR (95% CI) >1 y onlyc | 1.00 | 1.08 (0.70, 1.65) | 1.21 (0.77, 1.90) | 0.26 |
IL-10, pg/mL | <0.6 | 0.6–<1.6 | ≥1.6 | |
Cases/controls | 68/197 | 77/194 | 127/150 | |
OR (95% CI) all | 1.00 | 1.28 (0.85, 1.94) | 3.07 (2.02, 4.66) | <0.001 |
OR (95% CI) >1 y onlyc | 1.00 | 1.07 (0.67, 1.69) | 2.20 (1.38, 3.51) | 0.06 |
TNF-α, pg/mL | <1.7 | 1.7–<4.6 | ≥4.6 | |
Cases/controls | 95/177 | 81/191 | 96/173 | |
OR (95% CI) all | 1.00 | 0.78 (0.54, 1.14) | 1.04 (0.70, 1.54) | 0.05 |
OR (95% CI) >1 y onlyc | 1.00 | 0.73 (0.48, 1.11) | 0.98 (0.63, 1.54) | 0.23 |
CRP, mg/L | <1.0 | 1.1–<3.1 | ≥3.1 | |
Cases/controls | 92/183 | 85/178 | 94/178 | |
OR (95% CI) all | 1.00 | 0.94 (0.66, 1.36) | 1.05 (0.72, 1.52) | 0.32 |
OR (95% CI) >1 y onlyc | 1.00 | 0.78 (0.51, 1.18) | 0.85 (0.56, 1.31) | 0.70 |
Summary measurec | <−0.5 | −0.5–<0.1 | ≥0.1 | |
Cases/controls | 94/177 | 81/191 | 97/173 | |
OR (95% CI) all | 1.00 | 0.79 (0.53, 1.16) | 1.08 (0.73, 1.60) | 0.08 |
OR (95% CI) >1 y onlyc | 1.00 | 0.86 (0.55, 1.36) | 1.02 (0.65, 1.60) | 0.42 |
Abbreviations: BMI, body mass index; CRP, C-reactive protein IFN, Interferon; IL, Interleukin; TNF, Tumor neurosis factor
OR and 95% CI estimated by conditional logistic regression; cases and controls matched on sex, birthdate (± 1 year), ethnicity (white, Japanese American, Latino, African American, or Native Hawaiian), location (California or Hawaii), date of blood draw (within ±1 year), time of day of blood draw, and fasting hours prior to blood draw (0–<6, 6–<8, 8–<10, or ≥10). Missing data for leptin (1) and CRP (3).
p for trend based on the Wald χ2 test of a log-transformed continuous variable.
N=642 (219 cases and 423 controls)
Composite score from principal components analysis of log-transformed IL-1β, IL-4, IL-5, IL-6, IL-8, and TNF-α.
Adjustment for BMI had a limited effect. The inverse association between leptin and NHL risk was slightly strengthened (ORT3 vs. T1=0.30 [0.18–0.52]; ptrend<0.0001) and the positive association between IL-10 and NHL risk remained significant (ORT3 vs. T1=3.09 [2.03–4.70]; ptrend<0.001). BMI significantly modified the association of leptin with NHL (pinteraction<0.0001) but not of IL-10 (pinteraction =0.14) although the association was stronger in overweight than normal weight women (Table 3). There was no substantial evidence of effect modification by sex for leptin, IL-10, or IL-8 (pinteraction>0.23 for all). A weak interaction with ethnicity was seen only for IL-10 (pinteraction=0.11); the association was more pronounced in Latinos (ORT3 vs. T1= 4.50 [1.64, 12.38]; ptrend<0.01) and whites (ORT3 vs. T1= 6.74 [2.75, 16.54]; ptrend<0.01) than in the other groups (data not shown).
Table 3.
Body Mass Index (BMI)
|
pinteraction b | ||||
---|---|---|---|---|---|
<25 kg/m2 | 25 kg/m2 | ||||
| |||||
Cases/controls | OR (95% CI) | Cases/controls | OR (95% CI) | ||
Leptin, ng/ml | <0.0001 | ||||
<6.4 | 65/103 | 1.00 | 40/62 | 1.00 | |
6.4–<14.9 | 31/81 | 0.52 (0.24, 1.09) | 60/102 | 0.84 (0.45, 1.55) | |
≥14.9 | 13/41 | 0.35 (0.10, 1.18) | 63/151 | 0.54 (0.26, 1.12) | |
ptrend c | 0.02 | 0.02 | |||
IL-10, pg/mL | 0.14 | ||||
<0.64 | 28/83 | 1.00 | 40/114 | 1.00 | |
0.64–<1.19 | 33/77 | 0.82 (0.34, 1.93) | 44/117 | 0.87 (0.47, 1.62) | |
≥1.59 | 48/66 | 1.56 (0.65, 3.71) | 79/84 | 2.46 (1.33, 4.54) | |
ptrend c | 0.40 | <0.01 |
Abbreviations: BMI, body mass index; CI, Confidence interval; CRP, C-reactive protein; IFN, Interferon; IL, Interleukin; OR, odds ratio; TNF, Tumor neurosis factor.
OR and 95% CI estimated by conditional logistic regression; cases and controls matched on sex, birthdate (± 1 year), ethnicity (white, Japanese American, Latino, African American, or Native Hawaiian), location (California or Hawaii), date of blood draw (within ±1 year), time of day of blood draw, and fasting hours prior to blood draw (0–<6, 6–<8, 8–<10, or ≥10). Data were missing for leptin (1).
pinteraction based on the Wald χ2 test of the cross-product terms between log-transformed continuous serum measure and continuous BMI.
prend based on the Wald χ2 test of a log-transformed continuous variable.
Pre-diagnostic IL-10 or leptin levels were stronger predictors of NHL risk when the blood collection occurred closer to diagnosis (Table 4); however, a significant interaction with follow-up time was seen only for leptin (pinteraction<0.0001). For the 51 cases diagnosed >5 years after the blood draw, the estimated risk for the highest tertile of leptin was (ORT3 vs. T1=1.46 [0.48–4.45]; ptrend=0.80).
Table 4.
<2.7 years
|
2.7 years
|
pinteraction b | |||
---|---|---|---|---|---|
Cases/controls | OR (95% CI) | Cases/controls | OR (95% CI) | ||
Leptin, ng/ml | <0.0001 | ||||
<6.4 | 51/80 | 1.00 | 54/86 | 1.00 | |
6.4–<14.9 | 52/100 | 0.64 (0.36–1.15) | 39/82 | 0.69 (0.39–1.23) | |
≥14.9 | 33/97 | 0.38 (0.19–0.74) | 43/95 | 0.65 (0.33–1.27) | |
Ptrend c | <0.01 | 0.09 | |||
IL-10, pg/mL | |||||
<0.6 | 26/97 | 1.00 | 42/100 | 1.00 | 0.92 |
0.6<1.6 | 35/92 | 1.59 (0.83–3.06) | 42/102 | 0.95 (0.52–1.72) | |
≥1.6 | 75/89 | 3.88 (2.08–7.26) | 52/61 | 1.97 (1.07–3.62) | |
Ptrend c | <0.001 | 0.21 |
Abbreviations: BMI, body mass index; CI, Confidence interval; CRP, C-reactive protein IFN, Interferon; IL, Interleukin; OR, odds ratio; TNF, Tumor neurosis factor.
OR and 95% CI estimated by conditional logistic regression; cases and controls matched on sex, birthdate (± 1 year), ethnicity (white, Japanese American, Latino, African American, or Native Hawaiian), location (California or Hawaii), date of blood draw (within ±1 year), time of day of blood draw, and fasting hours prior to blood draw(0–<6, 6–<8, 8–<10, or ≥10). Data were missing for leptin (1).
pinteraction based on the Wald χ2 test of cross-product terms between the log-transformed continuous serum measure and years between blood draw and diagnosis.
ptrend based on the Wald χ2 test of a log-transformed continuous variable.
Although heterogeneity by NHL subtype was not statistically significant across markers, stratified analyses were performed for DLBCL and FL (Table 5). No significant associations were seen for DLBCL. However, just as in the overall analyses, leptin predicted a lower risk and IL-10 a higher risk for FL. In addition, IL-6 was associated with an elevated risk for FL. The exclusion of cases diagnosed <1 year after blood draw attenuated all risk estimates.
Table 5.
DLBCL | FL | |||||||
---|---|---|---|---|---|---|---|---|
| ||||||||
T1 | T2 | T3 | ptrend b | T1 | T2 | T3 | ptrend b | |
|
|
|||||||
Adiponectin, μg/mL | <5.6 | 5.6–<11.0 | ≥11.0 | <5.6 | 5.6–<11.0 | ≥11.0 | ||
Cases/controls | 28/49 | 24/57 | 27/51 | . | 16/32 | 19/30 | 14/34 | |
OR (95% CI) all | 1.00 | 0.70 (0.33, 1.45) | 0.85 (0.40, 1.78) | 0.83 | 1.00 | 1.19 (0.52, 2.73) | 0.82 (0.31, 2.20) | 0.80 |
OR (95% CI) >1 y onlyc | 1.00 | 0.61 (0.27, 1.39) | 0.86 (0.39, 1.92) | 0.84 | 1.00 | 1.06 (0.38, 2.99) | 0.50 (0.14, 1.75) | 0.35 |
Leptin, ng/ml | <6.4 | 6.4–<14.9 | ≥14.9 | <6.4 | 6.4–<14.9 | ≥14.9 | ||
Cases/controls | 28/45 | 28/56 | 23/56 | . | 17/21 | 20/34 | 12/41 | |
OR (95% CI) all | 1.00 | 0.73 (0.36, 1.50) | 0.49 (0.20, 1.24) | 0.33 | 1.00 | 0.49 (0.17, 1.38) | 0.13 (0.03, 0.53) | <0.01 |
OR (95% CI) >1 y onlyc | 1.00 | 0.96 (0.44, 2.13) | 0.74 (0.27, 2.00) | 0.93 | 1.00 | 0.49 (0.13, 1.78) | 0.16 (0.03, 0.95) | 0.06 |
IFN-γ, pg/mL | <0.5 | 0.5–<0.9 | ≥0.9 | <0.5 | 0.5–<0.9 | ≥0.9 | ||
Cases/controls | 24/52 | 26/64 | 29/41 | . | 20/35 | 12/28 | 17/33 | |
OR (95% CI) all | 1.00 | 0.91 (0.45, 1.82) | 1.60 (0.79, 3.26) | 0.23 | 1.00 | 0.72 (0.28, 1.82) | 0.85 (0.33, 2.16) | 0.93 |
OR (95% CI) >1 y onlyc | 1.00 | 0.76 (0.36, 1.60) | 1.41 (0.65, 3.06) | 0.64 | 1.00 | 0.53 (0.17, 1.70) | 0.91 (0.31, 2.68) | 0.88 |
IL-1β, pg/mL | <0.6 | 0.6–<1.6 | ≥1.6 | <0.6 | 0.6–<1.6 | ≥1.6 | ||
Cases/controls | 23/49 | 24/59 | 32/49 | . | 23/40 | 10/29 | 16/27 | |
OR (95% CI) all | 1.00 | 0.90 (0.43, 1.87) | 1.48 (0.72, 3.05) | 0.32 | 1.00 | 0.56 (0.21, 1.52) | 0.99 (0.38, 2.56) | 0.07 |
OR (95% CI) >1 y onlyc | 1.00 | 1.10 (0.50, 2.41) | 1.45 (0.64, 3.28) | 0.78 | 1.00 | 0.97 (0.32, 2.99) | 1.19 (0.39, 3.60) | 0.13 |
IL-2, pg/mL | <0.9 | 0.9–<1.9 | ≥1.9 | <0.9 | 0.9–<1.9 | ≥1.9 | ||
Cases/controls | 29/52 | 24/41 | 26/64 | . | 17/39 | 16/29 | 16/28 | |
OR (95% CI) all | 1.00 | 1.03 (0.49, 2.17) | 0.70 (0.34, 1.41) | 0.54 | 1.00 | 1.31 (0.53, 3.25) | 1.35 (0.53, 3.43) | 0.22 |
OR (95% CI) >1 y onlyc | 1.00 | 0.92 (0.40, 2.12) | 0.52 (0.23, 1.17) | 0.79 | 1.00 | 1.60 (0.53, 4.82) | 1.25 (0.38, 4.11) | 0.34 |
IL-4, pg/mL | <1.4 | 1.4–<3.4 | ≥3.4 | <1.4 | 1.4–<3.4 | ≥3.4 | ||
Cases/controls | 27/53 | 25/54 | 27/50 | . | 17/40 | 20/29 | 12/27 | |
OR (95% CI) all | 1.00 | 0.93 (0.48, 1.82) | 1.10 (0.50, 2.41) | 0.61 | 1.00 | 1.62 (0.67, 3.89) | 1.09 (0.40, 3.01) | 0.25 |
OR (95% CI) >1 y onlyc | 1.00 | 0.86 (0.41, 1.77) | 0.96 (0.40, 2.35) | 0.31 | 1.00 | 1.92 (0.68, 5.45) | 1.18 (0.36, 3.83) | 0.21 |
IL-5, pg/mL | <0.4 | 0.4–<0.9 | ≥0.9 | <0.4 | 0.4–<0.9 | ≥0.9 | ||
Cases/controls | 18/50 | 30/51 | 31/56 | . | 17/43 | 16/27 | 16/26 | |
OR (95% CI) all | 1 | 1.84 (0.85, 3.99) | 1.79 (0.80, 4.01) | 0.92 | 1 | 1.67 (0.71, 3.92) | 2.12 (0.73, 6.13) | 0.05 |
OR (95% CI) >1 y onlyc | 1 | 2.12 (0.88, 5.12) | 2.17 (0.88, 5.32) | 0.86 | 1 | 1.49 (0.52, 4.26) | 1.45 (0.36, 5.80) | 0.15 |
IL-6, pg/mL | <1.1 | 1.1–<2.7 | ≥2.7 | <1.1 | 1.1–<2.7 | ≥2.7 | ||
Cases/controls | 25/54 | 28/55 | 26/48 | . | 14/40 | 19/33 | 16/23 | |
OR (95% CI) all | 1.00 | 1.13 (0.58, 2.20) | 1.20 (0.58, 2.48) | 0.73 | 1.00 | 1.71 (0.72, 4.07) | 2.05 (0.81, 5.13) | 0.04 |
OR (95% CI) >1 y onlyc | 1.00 | 1.17 (0.57, 2.42) | 0.82 (0.35, 1.94) | 0.54 | 1.00 | 1.26 (0.44, 3.57) | 2.42 (0.75, 7.80) | 0.24 |
IL-8, pg/mL | <6.7 | 6.7–<14.6 | ≥14.6 | <6.7 | 6.7–<14.6 | ≥14.6 | ||
Cases/controls | 20/41 | 29/63 | 30/53 | . | 16/41 | 16/27 | 17/28 | |
OR (95% CI) all | 1.00 | 0.93 (0.45, 1.93) | 1.15 (0.56, 2.36) | 0.40 | 1.00 | 1.61 (0.65, 3.95) | 1.70 (0.66, 4.35) | 0.13 |
OR (95% CI) >1 y onlyc | 1.00 | 0.96 (0.45, 2.07) | 1.04 (0.48, 2.26) | 0.62 | 1.00 | 1.70 (0.56, 5.20) | 2.92 (0.83, | 0.13 |
IL-10, pg/mL | <0.6 | 0.6–<1.6 | ≥1.6 | <0.6 | 0.6–<1.6 | ≥1.6 | ||
Cases/controls | 24/56 | 20/58 | 35/43 | . | 9/46 | 14/29 | 26/21 | |
OR (95% CI) all | 1.00 | 0.80 (0.39, 1.66) | 2.36 (1.10, 5.05) | 0.06 | 1.00 | 4.15 (1.22, | 8.46 (2.71, | 0.01 |
OR (95% CI) >1 y onlyc | 1.00 | 0.61 (0.28, 1.33) | 1.62 (0.71, 3.69) | 0.64 | 1.00 | 5.11 (1.23, | 7.44 (1.97, | 0.12 |
TNF-α, pg/mL | <1.7 | 1.7–<4.6 | ≥4.6 | <1.7 | 1.7–<4.6 | ≥4.6 | ||
Cases/controls | 32/42 | 23/66 | 24/49 | . | 17/35 | 15/27 | 17/34 | |
OR (95% CI) all | 1.00 | 0.42 (0.21, 0.85) | 0.58 (0.28, 1.22) | 0.92 | 1.00 | 1.16 (0.46, 2.92) | 1.03 (0.41, 2.61) | 0.28 |
OR (95% CI) >1 y onlyc | 1.00 | 0.40 (0.19, 0.84) | 0.68 (0.31, 1.51) | 0.68 | 1.00 | 1.09 (0.36, 3.29) | 0.85 (0.28, 2.56) | 0.31 |
CRP, mg/L | <1.0 | 1.1–<3.1 | ≥3.1 | <1.0 | 1.1–<3.1 | ≥3.1 | ||
Cases/controls | 30/54 | 22/52 | 27/51 | . | 16/32 | 17/32 | 16/32 | |
OR (95% CI) all | 1.00 | 0.78 (0.41, 1.50) | 0.97 (0.49, 1.92) | 0.26 | 1.00 | 1.03 (0.43, 2.47) | 0.96 (0.38, 2.43) | 0.64 |
OR (95% CI) >1 y onlyc | 1.00 | 0.54 (0.26, 1.13) | 0.64 (0.30, 1.40) | 0.78 | 1.00 | 0.97 (0.31, 3.01) | 0.85 (0.26, 2.74) | 0.84 |
Summary measured | <−0.5 | −0.5–<0.1 | ≥0.1 | <−0.5 | −0.5–<0.1 | ≥0.1 | ||
Cases/controls | 24/53 | 28/54 | 27/50 | . | 20/42 | 14/25 | 15/29 | |
OR (95% CI) all | 1.00 | 1.15 (0.59, 2.25) | 1.23 (0.60, 2.50) | 0.60 | 1.00 | 1.22 (0.51, 2.92) | 1.09 (0.45, 2.62) | 0.10 |
OR (95% CI) >1 y onlyc | 1.00 | 0.78 (0.38, 1.59) | 1.04 (0.46, 2.33) | 0.95 | 1.00 | 1.11 (0.37, 3.28) | 1.04 (0.36, 3.01) | 0.17 |
Abbrev: DLBCL, diffuse large B-cell lymphoma; BMI, body mass index; CRP, C-reactive protein IFN, Interferon; IL, Interleukin; TNF, Tumor neurosis factor.
OR and 95% CI estimated by conditional logistic regression; cases and controls matched on sex, birthdate (± 1 year), ethnicity, location (California or Hawaii), date of blood draw (within ±1 year), time of day of blood draw, and fasting hours prior to blood draw (0–<6, 6–<8, 8–<10, or ≥10).
prend based on the Wald χ2 test of a log-transformed continuous variable.
N=198 (66 cases and 132 controls) for DLBCL and N=97 (33 cases and 64 controls) for follicular lymphoma
Composite score from principal components analysis of log-transformed IL-1β, IL-4, IL-5, IL-6, IL-8, and TNF-α.
Discussion
In this ethnically diverse nested case-control study, the highest tertile of pre-diagnostic, serum IL-10 levels was associated with a 3-fold elevation in NHL risk, while leptin was associated with a 50% reduction in NHL risk. Given the short follow-up and the attenuated risk estimates after exclusion of cases diagnosed <1 year after blood draw, these associations could be due to pre-clinical effects of the disease. Adjustment for BMI had little effect, but after stratification by NHL subtype, significant associations were restricted to FL. None of the other cytokines, CRP, and adiponectin showed a significant association with NHL risk.
The positive association with IL-10 agrees with one of the three published reports to date (20–22). In contrast to null reports from the New York University Women’s Health Study with 92 cases and 184 controls (21) and the Italian component of the European Prospective Investigation into Cancer and Nutrition (EPIC) study with 86 cases and controls each (22), a nested case-control study within the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial (297 cases and 297 matched controls) (20) observed a 2-fold higher NHL risk for the top versus the bottom quartile of circulating IL-10 (>7.77 vs. ≤1.93 pg/ml). Consistent with our findings in Table 4 and our results after excluding cases during the first year, the association was attenuated for cases diagnosed 2–5 years after the blood draw and not significant for cases diagnosed 6–10 years post-collection suggesting early tumor-induced effects. None of the three previous analyses reported separate risk estimates for DLBCL and FL (20–22).
On the other hand, two of the previous studies found a positive association with TNF-α (20, 21) and one found an inverse relation (22), whereas the current study found a borderline significant p-value for trend that disappeared after excluding cases diagnosed within 1 year of the blood draw. In the PLCO study (20), the highest versus the lowest quartile for circulating TNF-α (>7.98 vs. ≤3.96 pg/ml) levels was associated with a 2-fold higher risk and in the New York University Women’s Health Study, higher TNF-α and lower IL-5 levels were marginally associated with the risk for B-cell lymphoma (21). The Italian EPIC study (22) reported a 50% lower risk for higher TNF-α, but the 95% CI included one (ptrend=0.10). The inverse association between TNF-α and NHL only reached statistical significance when the analysis was restricted to B-cell lymphomas. The significant inverse associations of IL-2 and IFN-γ with NHL risk in overall models and after excluding cases diagnosed within 2 years of blood donation in the EPIC study (22) also disagree with the null findings observed in the current study.
The role of IL-10 in NHL pathogenesis is unclear. This pleiotropic cytokine with immunosuppressive and anti-inflammatory effects, as well as immunostimulatory effects on B-cells (37), may promote lymphomagenesis through the proliferation of B-cells and, thereby increase the likelihood of DNA-modifications, such as chromosomal translocations and oncogene mutations (38). In vivo studies of IL-10 knockout mice demonstrated the requirement for IL-10 in the progression of B-cell lymphoma (39). Elevated IL-10 levels have also been observed in NHL cases with poor prognosis (40–42) and in relation to the development of AIDS-related B-cell lymphoma (43). The association of pre-diagnostic serum IL-10 with NHL risk in the current study was observed with a relatively short lag-time between blood donation and diagnosis of 2.7 years. Therefore, reverse causation due to early tumor changes cannot be excluded given that malignant NHL cells produce IL-10 (44, 45).
To our knowledge, this was the first study to prospectively examine the association between circulating leptin and adiponectin levels and subsequent NHL risk. The strong inverse association between leptin and NHL risk observed in the present study appears to be inconsistent with the a priori hypothesis that leptin may mediate the positive obesity-NHL association (46). Moreover, the NHL risk estimates for increasing tertile levels of leptin were more pronounced after adjustment for BMI but did not differ substantially by BMI category (<25 vs. ≥25 kg/m2). Leptin is predominately produced by adipose tissue in proportion to body fat mass (47) and plays a central role in regulating energetic homeostasis (48). Recently, data from animal models and human studies have demonstrated the role of leptin in immune homeostasis and the innate and adaptive immune response, as reviewed elsewhere (27, 49). The leptin signaling receptor, OB-Rb, is found on various immune cells, e.g., monocytes, macrophages, NK cells, B-cells, T-cells, regulatory T-cells (49), and genetic polymorphisms in the leptin gene and its receptor support a role for leptin in NHL etiology (19, 46). However, the inverse relation between leptin and NHL risk is difficult to explain and the lack of a biological explanation suggests caution in the interpretation. Through its immunomodulatory effect, leptin may enhance the immune response, resulting in a more effective immunosurveillance by promoting NK-cell cytotoxicity and a switch to Th1-cell immune response (27). In regard to adiponectin, although the present study observed no association between adiponectin levels and NHL risk, adiponectin was higher in newly diagnosed NHL patients (n=28) than in age- and sex-matched controls (n=17) in a small observational study (41). Considering the current clinical and observational data supporting the role of adiponectin in cancer development and prognosis (50), including adiponectin receptor expression in NHL tissue samples (51) and in human lymphocytes (52), further studies are needed to investigate the potential role of adiponectin in NHL pathogenesis.
This study had several strengths including the prospective design that allowed for the pre-diagnostic assessment of biomarkers, the ethnic diversity of the study sample, the population-based sampling frame allowing for generalizability of results, and the principal components analysis that made use of multiple inflammatory marker measures. Although NHL diagnoses could not be confirmed by a re-review of pathology records, the majority of histologic subtypes are expected to be correctly classified within the SEER registry; all cases were diagnosed after the International Classification of Diseases for Oncology, Third Edition, had been adopted (1). The standardized blood collection and processing procedures minimized systematic error and variation in serum marker concentrations; all assays were performed in duplicate.
There are also a number of limitations. The main ones are the availability of a single blood sample for the analysis and the short time period between blood donation and NHL diagnosis (median 2.7 years). Therefore, the results from this study could be the result of reverse causation, i.e., the higher IL-10 and the lower leptin levels may indicate early changes due to lymphoma development as indicated by the lower risk estimates after the exclusion of early cases. Longer follow-up of the MEC and other cohorts is needed to confirm these findings. Although information on HIV status, family history of lymphoma, presence of autoimmune disease, immunosuppression, or agricultural exposure was not available and could not be examined as potential confounders, these conditions are expected to be rare in this relatively healthy cohort population. In addition, although the overall number of cases was larger than in previous reports, we had limited ability to detect statistically significant associations in subgroup analyses (20–22).
In conclusion, our findings provide little support for the hypothesis that pre-diagnostic levels of circulating immune markers play a role in the development of NHL. The associations of IL-10 and leptin with NHL risk should be interpreted with caution due to the potential for reverse causation and a lack of a clear biological mechanism. Additional prospective studies with repeated measures over a longer pre-diagnostic time period are needed to provide insight into the etiologic role of subclinical variations in immune markers in NHL pathogenesis.
Acknowledgments
The Multiethnic Cohort Study has been supported by National Cancer Institute grant R37 CA 54281 (PI: Dr. Kolonel) and the biorepository by P01 CA 033619 (PI: Dr. Kolonel). SMC was supported by a postdoctoral fellowship on grant R25 CA 90956. The tumor registries in Hawaii and Los Angeles are supported by NCI contracts N01 PC 35137 and N01 PC 35139. We thank Elysse Tom for her contribution to the data analysis and William Cooney and Jennifer Lai for their work on the laboratory ELISA and Luminex assays.
Footnotes
Conflict of Interest: None of the authors has a conflict of interest to declare.
References
- 1.Turner JJ, Morton LM, Linet MS, Clarke CA, Kadin ME, Vajdic CM, et al. InterLymph hierarchical classification of lymphoid neoplasms for epidemiologic research based on the WHO classification (2008): update and future directions. Blood. 2010;116:e90–e98. doi: 10.1182/blood-2010-06-289561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Engels EA. Infectious agents as causes of non-Hodgkin lymphoma. Cancer Epidemiol Biomarkers Prev. 2007;16:401–4. doi: 10.1158/1055-9965.EPI-06-1056. [DOI] [PubMed] [Google Scholar]
- 3.Grulich AE, Vajdic CM, Cozen W. Altered immunity as a risk factor for non-Hodgkin lymphoma. Cancer Epidemiol Biomarkers Prev. 2007;16:405–8. doi: 10.1158/1055-9965.EPI-06-1070. [DOI] [PubMed] [Google Scholar]
- 4.Zintzaras E, Voulgarelis M, Moutsopoulos HM. The risk of lymphoma development in autoimmune diseases: a meta-analysis. Arch Intern Med. 2005;165:2337–44. doi: 10.1001/archinte.165.20.2337. [DOI] [PubMed] [Google Scholar]
- 5.Ekstrom SK, Vajdic CM, Falster M, Engels EA, Martinez-Maza O, Turner J, et al. Autoimmune disorders and risk of non-Hodgkin lymphoma subtypes: a pooled analysis within the InterLymph Consortium. Blood. 2008;111:4029–38. doi: 10.1182/blood-2007-10-119974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Vendrame E, Martinez-Maza O. Assessment of pre-diagnosis biomarkers of immune activation and inflammation: insights on the etiology of lymphoma. J Proteome Res. 2011;10:113–9. doi: 10.1021/pr100729z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chiu BC, Weisenburger DD. An update of the epidemiology of non-Hodgkin’s lymphoma. Clin Lymphoma. 2003;4:161–8. doi: 10.3816/clm.2003.n.025. [DOI] [PubMed] [Google Scholar]
- 8.Mori T, Takada R, Watanabe R, Okamoto S, Ikeda Y. T-helper (Th)1/Th2 imbalance in patients with previously untreated B-cell diffuse large cell lymphoma. Cancer Immunol Immunother. 2001;50:566–8. doi: 10.1007/s00262-001-0232-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Lucey DR, Clerici M, Shearer GM. Type 1 and type 2 cytokine dysregulation in human infectious, neoplastic, and inflammatory diseases. Clin Microbiol Rev. 1996;9:532–62. doi: 10.1128/cmr.9.4.532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Eisenstein EM, Williams CB. The T(reg)/Th17 cell balance: a new paradigm for autoimmunity. Pediatr Res. 2009;65:26R–31R. doi: 10.1203/PDR.0b013e31819e76c7. [DOI] [PubMed] [Google Scholar]
- 11.Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441:235–8. doi: 10.1038/nature04753. [DOI] [PubMed] [Google Scholar]
- 12.Yang ZZ, Novak AJ, Ziesmer SC, Witzig TE, Ansell SM. Malignant B cells skew the balance of regulatory T cells and TH17 cells in B-cell non-Hodgkin’s lymphoma. Cancer Res. 2009;69:5522–30. doi: 10.1158/0008-5472.CAN-09-0266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Bel H, Jr, Chatti A, Laatiri A, Ahmed SB, Romdhane A, Ajimi S, et al. Tumor necrosis factor promoter gene polymorphism associated with increased susceptibility to non-Hodgkin’s lymphomas. Eur J Haematol. 2007;78:117–22. doi: 10.1111/j.1600-0609.2006.00784.x. [DOI] [PubMed] [Google Scholar]
- 14.Chen Y, Zheng T, Lan Q, Foss F, Kim C, Chen X, et al. Cytokine polymorphisms in Th1/Th2 pathway genes, body mass index, and risk of non-Hodgkin lymphoma. Blood. 2011;117:585–90. doi: 10.1182/blood-2010-07-295097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lan Q, Zheng T, Rothman N, Zhang Y, Wang SS, Shen M, et al. Cytokine polymorphisms in the Th1/Th2 pathway and susceptibility to non-Hodgkin lymphoma. Blood. 2006;107:4101–8. doi: 10.1182/blood-2005-10-4160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Purdue MP, Lan Q, Kricker A, Grulich AE, Vajdic CM, Turner J, et al. Polymorphisms in immune function genes and risk of non-Hodgkin lymphoma: findings from the New South Wales non-Hodgkin Lymphoma Study. Carcinogenesis. 2007;28:704–12. doi: 10.1093/carcin/bgl200. [DOI] [PubMed] [Google Scholar]
- 17.Rothman N, Skibola CF, Wang SS, Morgan G, Lan Q, Smith MT, et al. Genetic variation in TNF and IL10 and risk of non-Hodgkin lymphoma: a report from the InterLymph Consortium. Lancet Oncol. 2006;7:27–38. doi: 10.1016/S1470-2045(05)70434-4. [DOI] [PubMed] [Google Scholar]
- 18.Wang SS, Cerhan JR, Hartge P, Davis S, Cozen W, Severson RK, et al. Common genetic variants in proinflammatory and other immunoregulatory genes and risk for non-Hodgkin lymphoma. Cancer Res. 2006;66:9771–80. doi: 10.1158/0008-5472.CAN-06-0324. [DOI] [PubMed] [Google Scholar]
- 19.Willett EV, Skibola CF, Adamson P, Skibola DR, Morgan GJ, Smith MT, et al. Non-Hodgkin’s lymphoma, obesity and energy homeostasis polymorphisms. Br J Cancer. 2005;93:811–6. doi: 10.1038/sj.bjc.6602762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Purdue MP, Lan Q, Bagni R, Hocking WG, Baris D, Reding DJ, et al. Prediagnostic serum levels of cytokines and other immune markers and risk of non-hodgkin lymphoma. Cancer Res. 2011;71:4898–907. doi: 10.1158/0008-5472.CAN-11-0165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Gu Y, Shore RE, Arslan AA, Koenig KL, Liu M, Ibrahim S, et al. Circulating cytokines and risk of B-cell non-Hodgkin lymphoma: a prospective study. Cancer Causes Control. 2010;21:1323–33. doi: 10.1007/s10552-010-9560-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Saberi HF, Krop EJ, Scoccianti C, Krogh V, Palli D, Panico S, et al. Plasma cytokines and future risk of non-Hodgkin lymphoma (NHL): a case-control study nested in the Italian European Prospective Investigation into Cancer and Nutrition. Cancer Epidemiol Biomarkers Prev. 2010;19:1577–84. doi: 10.1158/1055-9965.EPI-09-1237. [DOI] [PubMed] [Google Scholar]
- 23.Marti A, Marcos A, Martinez JA. Obesity and immune function relationships. Obes Rev. 2001;2:131–40. doi: 10.1046/j.1467-789x.2001.00025.x. [DOI] [PubMed] [Google Scholar]
- 24.Larsson SC, Wolk A. Obesity and risk of non-Hodgkin’s lymphoma: a meta-analysis. Int J Cancer. 2007;121:1564–70. doi: 10.1002/ijc.22762. [DOI] [PubMed] [Google Scholar]
- 25.Willett EV, Morton LM, Hartge P, Becker N, Bernstein L, Boffetta P, et al. Non-Hodgkin lymphoma and obesity: a pooled analysis from the InterLymph Consortium. Int J Cancer. 2008;122:2062–70. doi: 10.1002/ijc.23344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Larsson SC, Wolk A. Body mass index and risk of non-Hodgkin’s and Hodgkin’s lymphoma: a meta-analysis of prospective studies. Eur J Cancer. 2011;47:2422–30. doi: 10.1016/j.ejca.2011.06.029. [DOI] [PubMed] [Google Scholar]
- 27.La CA, Matarese G. The weight of leptin in immunity. Nat Rev Immunol. 2004;4:371–9. doi: 10.1038/nri1350. [DOI] [PubMed] [Google Scholar]
- 28.Lam QL, Wang S, Ko OK, Kincade PW, Lu L. Leptin signaling maintains B-cell homeostasis via induction of Bcl-2 and Cyclin D1. Proc Natl Acad Sci U S A. 2010;107:13812–7. doi: 10.1073/pnas.1004185107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Yokota T, Oritani K, Takahashi I, Ishikawa J, Matsuyama A, Ouchi N, et al. Adiponectin, a new member of the family of soluble defense collagens, negatively regulates the growth of myelomonocytic progenitors and the functions of macrophages. Blood. 2000;96:1723–32. [PubMed] [Google Scholar]
- 30.Wolf AM, Wolf D, Rumpold H, Enrich B, Tilg H. Adiponectin induces the anti-inflammatory cytokines IL-10 and IL-1RA in human leukocytes. Biochem Biophys Res Commun. 2004;323:630–5. doi: 10.1016/j.bbrc.2004.08.145. [DOI] [PubMed] [Google Scholar]
- 31.Karin M, Greten FR. NF-kappaB: linking inflammation and immunity to cancer development and progression. Nat Rev Immunol. 2005;5:749–59. doi: 10.1038/nri1703. [DOI] [PubMed] [Google Scholar]
- 32.Wang SS, Cozen W, Cerhan JR, Colt JS, Morton LM, Engels EA, et al. Immune mechanisms in non-Hodgkin lymphoma: joint effects of the TNF G308A and IL10 T3575A polymorphisms with non-Hodgkin lymphoma risk factors. Cancer Res. 2007;67:5042–54. doi: 10.1158/0008-5472.CAN-06-4752. [DOI] [PubMed] [Google Scholar]
- 33.Kolonel LN, Henderson BE, Hankin JH, Nomura AM, Wilkens LR, Pike MC, et al. A multiethnic cohort in Hawaii and Los Angeles: baseline characteristics. Am J Epidemiol. 2000;151:346–57. doi: 10.1093/oxfordjournals.aje.a010213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Park SY, Wilkens LR, Henning SM, Le ML, Gao K, Goodman MT, et al. Circulating fatty acids and prostate cancer risk in a nested case-control study: the Multiethnic Cohort. Cancer Causes Control. 2009;20:211–23. doi: 10.1007/s10552-008-9236-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hatcher L. A step-by-step approach to using the SAS system for factor analysis and structural equation modeling. Cary, NC: SAS Publishing; 1994. [Google Scholar]
- 36.Mickey RM, Greenland S. The impact of confounder selection criteria on effect estimation. Am J Epidemiol. 1989;129:125–37. doi: 10.1093/oxfordjournals.aje.a115101. [DOI] [PubMed] [Google Scholar]
- 37.Mocellin S, Marincola F, Rossi CR, Nitti D, Lise M. The multifaceted relationship between IL-10 and adaptive immunity: putting together the pieces of a puzzle. Cytokine Growth Factor Rev. 2004;15:61–76. doi: 10.1016/j.cytogfr.2003.11.001. [DOI] [PubMed] [Google Scholar]
- 38.Epeldegui M, Widney DP, Martinez-Maza O. Pathogenesis of AIDS lymphoma: role of oncogenic viruses and B cell activation-associated molecular lesions. Curr Opin Oncol. 2006;18:444–8. doi: 10.1097/01.cco.0000239882.23839.e5. [DOI] [PubMed] [Google Scholar]
- 39.Czarneski J, Lin YC, Chong S, McCarthy B, Fernandes H, Parker G, et al. Studies in NZB IL-10 knockout mice of the requirement of IL-10 for progression of B-cell lymphoma. Leukemia. 2004;18:597–606. doi: 10.1038/sj.leu.2403244. [DOI] [PubMed] [Google Scholar]
- 40.Guney N, Soydinc HO, Basaran M, Bavbek S, Derin D, Camlica H, et al. Serum levels of interleukin-6 and interleukin-10 in Turkish patients with aggressive non-Hodgkin’s lymphoma. Asian Pac J Cancer Prev. 2009;10:669–74. [PubMed] [Google Scholar]
- 41.Pamuk GE, Turgut B, Demir M, Vural O. Increased adiponectin level in non-Hodgkin lymphoma and its relationship with interleukin-10. Correlation with clinical features and outcome. J Exp Clin Cancer Res. 2006;25:537–41. [PubMed] [Google Scholar]
- 42.Lech-Maranda E, Bienvenu J, Broussais-Guillaumot F, Warzocha K, Michallet AS, Robak T, et al. Plasma TNF-alpha and IL-10 level-based prognostic model predicts outcome of patients with diffuse large B-Cell lymphoma in different risk groups defined by the International Prognostic Index. Arch Immunol Ther Exp (Warsz ) 2010;58:131–41. doi: 10.1007/s00005-010-0066-1. [DOI] [PubMed] [Google Scholar]
- 43.Breen EC, Boscardin WJ, Detels R, Jacobson LP, Smith MW, O’Brien SJ, et al. Non-Hodgkin’s B cell lymphoma in persons with acquired immunodeficiency syndrome is associated with increased serum levels of IL10, or the IL10 promoter -592 C/C genotype. Clin Immunol. 2003;109:119–29. doi: 10.1016/s1521-6616(03)00214-6. [DOI] [PubMed] [Google Scholar]
- 44.Emilie D, Touitou R, Raphael M, Peuchmaur M, Devergnee O, Rea D, et al. In vivo production of interleukin-10 by malignant cells in AIDS lymphomas. Eur J Immunol. 1992;22:2937–42. doi: 10.1002/eji.1830221127. [DOI] [PubMed] [Google Scholar]
- 45.Voorzanger N, Touitou R, Garcia E, Delecluse HJ, Rousset F, Joab I, et al. Interleukin (IL)-10 and IL-6 are produced in vivo by non-Hodgkin’s lymphoma cells and act as cooperative growth factors. Cancer Res. 1996;56:5499–505. [PubMed] [Google Scholar]
- 46.Skibola CF, Holly EA, Forrest MS, Hubbard A, Bracci PM, Skibola DR, et al. Body mass index, leptin and leptin receptor polymorphisms, and non-hodgkin lymphoma. Cancer Epidemiol Biomarkers Prev. 2004;13:779–86. [PubMed] [Google Scholar]
- 47.Thomas T, Burguera B, Melton LJ, III, Atkinson EJ, O’Fallon WM, Riggs BL, et al. Relationship of serum leptin levels with body composition and sex steroid and insulin levels in men and women. Metabolism. 2000;49:1278–84. doi: 10.1053/meta.2000.9519. [DOI] [PubMed] [Google Scholar]
- 48.Friedman JM, Halaas JL. Leptin and the regulation of body weight in mammals. Nature. 1998;395:763–70. doi: 10.1038/27376. [DOI] [PubMed] [Google Scholar]
- 49.Procaccini C, Jirillo E, Matarese G. Leptin as an immunomodulator. Mol Aspects Med. 2012;33:35–45. doi: 10.1016/j.mam.2011.10.012. [DOI] [PubMed] [Google Scholar]
- 50.Dalamaga M, Diakopoulos KN, Mantzoros CS. The role of adiponectin in cancer: a review of current evidence. Endocr Rev. 2012;33:547–94. doi: 10.1210/er.2011-1015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Petridou ET, Sergentanis TN, Dessypris N, Vlachantoni IT, Tseleni-Balafouta S, Pourtsidis A, et al. Serum adiponectin as a predictor of childhood non-Hodgkin’s lymphoma: a nationwide case-control study. J Clin Oncol. 2009;27:5049–55. doi: 10.1200/JCO.2008.19.7525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Alberti L, Gilardini L, Girola A, Moro M, Cavagnini F, Invitti C. Adiponectin receptors gene expression in lymphocytes of obese and anorexic patients. Diabetes Obes Metab. 2007;9:344–9. doi: 10.1111/j.1463-1326.2006.00614.x. [DOI] [PubMed] [Google Scholar]