Smaller-scale evaluations suggest that common genetic variation in candidate genes related to immune function may predispose to the development of non-Hodgkin lymphoma (NHL). We report an analysis of variants within genes associated with immunity and inflammation and risk of NHL using a panel of 9412 single-nucleotide polymorphisms (SNPs) from 1253 genes in a study of 458 patients with NHL and 484 frequency-matched controls. We modeled haplotypes and risk of NHL, as well as the main effects for all independent SNPs from a gene in multivariate logistic regression models; we separately report results for nonsynonymous (ns) SNPs. In gene-level analyses, the strongest findings (P ≤ .001) were for CREB1, FGG, MAP3K5, RIPK3, LSP1, TRAF1, DUSP2, and ITGB3. In nsSNP analyses, the strongest findings (P ≤ .01) were for ITGB3 L59P (odds ratio [OR] = 0.66; 95% confidence interval [CI] 0.52-0.85), TLR6 V427A (OR = 5.20; CI 1.77-15.3), SELPLG M264V (OR = 3.20; CI 1.48-6.91), UNC84B G671S (OR = 1.50; CI 1.12-2.00), B3GNT3 H328R (OR = 0.74; CI 0.59-0.93), and BAT2 V1883L (OR = 0.64; CI 0.45-0.90). Our results suggest that genetic variation in genes associated with immune response (TRAF1, RIPK3, BAT2, and TLR6), mitogen-activated protein kinase (MAPK) signaling (MAP3K5, DUSP2, and CREB1), lymphocyte trafficking and migration (B3GNT3, SELPLG, and LSP1), and coagulation pathways (FGG and ITGB3) may be important in the etiology of NHL, and should be prioritized in replication studies.

Non-Hodgkin lymphoma (NHL) is the most commonly diagnosed hematologic malignancy in the United States,1  and the lifetime odds of developing NHL is 1 in 47 for men and 1 in 55 for women.2  Given the remarkable rise in incidence of NHL in the last 50 years, it is clear that environmental factors must play a major role in the etiology of this cancer, although established risk factors to date account for only a relatively small fraction of the total number of cases.3  There is also accumulating evidence from migrant and analytic epidemiology studies that genetic susceptibility plays a role in NHL etiology,4,5  although to date no major gene has been identified. However, case-control studies have identified several promising candidate susceptibility genes supporting a polygenic model based on low-penetrance alleles in line with the common-variant, common-disease hypothesis.6 

For NHL, the most compelling hypothesis for increased cancer risk is immune dysfunction, and this risk may be influenced in part by variation in polymorphic genes that control immune function and regulation. Recent studies have shown that single-nucleotide polymorphisms (SNPs) from candidate genes, including TNF and LTA7,9 ; several interleukin genes, including IL4, IL5, IL6, and IL108,10,11 ; and genes related to innate immunity, including FCGR2A,9 TLR4,12  and CARD15,12  may be risk factors for NHL overall or for certain NHL subtypes.

We undertook a discovery exercise to identify additional genes related to immune function and inflammation and risk of NHL by using the Affymetrix Immune and Inflammation SNP panel, which included 9412 SNPs from 1253 genes. SNP coverage included both nonsynonymous (ns) SNPs (N = 537) and tagSNPs derived from the HapMap. Our analyses focused on both a gene-level test using all of the SNPs, and for nsSNPs a SNP-level test, as the latter SNPs are expected to have a higher prior probability of functional significance. The study was conducted using a clinic-based case-control study of 458 patients with NHL and 484 frequency-matched controls, and was based at the Mayo Clinic (Rochester, MN).

Study population

This study was reviewed and approved by the Human Subjects Institutional Review Board at the Mayo Clinic, and informed consent was obtained in accordance with the Declaration of Helsinki from all participants. All consecutive patients with histologically confirmed lymphoma (Hodgkin lymphoma [HL] and NHL, including chronic lymphocytic leukemia [CLL]) aged 20 years and older, who were residents of Minnesota, Iowa, or Wisconsin at the time of diagnosis, and who were within 9 months of their initial diagnosis at presentation to Mayo Clinic Rochester from September 1, 2002, forward were offered enrollment into the study. Patients were excluded if they had HIV infection, did not speak English, or were unable to provide written informed consent. A Mayo hematopathologist reviewed all materials for each case to verify the diagnosis and to classify each case into the World Health Organization Classification of Neoplastic Diseases of the Hematopoietic and Lymphoid Tissues.13  This (phase 1) analysis included all patients enrolled into the study from September 1, 2002, through September 30, 2005. Of the 956 eligible patients identified during this time frame, 629 (66%) participated, 106 (11%) refused, 19 (2%) were unable to be contacted, and 202 (24%) had their eligibility expire (ie, after identification they did not consent within 9 months of diagnosis, or after consent they did not complete data collection within 12 months of diagnosis). Most patients who had their eligibility expire were only seen once at the Mayo Clinic for a second opinion, and these patients were generally recruited through the mail rather than in person.

Clinic-based controls were randomly selected from Mayo Clinic Rochester patients aged 20 years and older, who were residents of Minnesota, Iowa, or Wisconsin, and who were being seen for a prescheduled medical examination in the general medicine divisions of the Department of Medicine from September 1, 2002, to September 30, 2005. Patients were not eligible if they had a history of lymphoma or leukemia, had HIV infection, or did not speak English. Controls were frequency-matched to the case distribution on 5-year age group, sex, and county of residence (county groupings based on distance from Rochester and urban/rural status) using a computer program that randomly selects subjects from eligible patients. Of the 818 eligible subjects identified, 572 (70%) participated, 239 (29%) refused, and 7 (1%) had their eligibility expire (ie, did not complete data collection within 12 months of selection).

Data collection

Participants completed a risk-factor questionnaire that included data on demographics, ethnicity, family cancer history, medical history, and selected lifestyle factors; they also provided a peripheral blood sample for serologic and genetic studies. DNA was extracted from samples using a Gentra Systems automated salting-out methodology (Gentra, Minneapolis, MN). A total of 498 (79%) patients and 497 (87%) controls had an extracted DNA sample available for genotyping in November 2005. DNA samples were randomly assigned to 1 of 12 96-well plates. We also randomly selected (without replacement) 2 samples that were duplicated across plates and another 2 samples that were duplicated within each plate, for a total of 1043 study samples (995 unique samples and 48 duplicates). In addition, 8 wells on each plate were left blank for additional quality control samples (N = 96). The remaining wells were either blank (N = 10) or had water only (N = 3). All source DNA tubes were barcoded and wanded into the Biomek NX software system (Beckman Coulter, Fullerton, CA), creating a virtual plate map of the robotically plated samples.

Genotyping and quality control

Genotyping was conducted at the Affymetrix facility in South San Francisco, CA, using the Molecular Inversion Probe (MIP) genotyping technology,14  which has a robust genotype-calling methodology.15  The 9K Immune-Inflammation Panel consists of 9412 MIP assays representing SNPs in 1253 genes selected for their involvement in inflammation and immunity. HapMap data (http://www.hapmo-p.org, phase 1, version 16) from CEPH (Centre d'Etude du Polymorphisme Humain; white) and Yoruba (African) samples were used to select tagging SNPs, and these SNPs were chosen to give an r2 coverage of 0.8 or greater for all SNPs genotyped in the HapMap that had a minor allele frequency (MAF) of more than 5%. These SNPs covered the entire gene, from 5 kb upstream to 5 kb downstream of the gene, as well as all exons and introns. In addition, the panel included 748 validated nsSNPs. The average number of SNPs per gene was 6.8. A complete list of the SNPs on the assay panel is available from the Affymetrix website (http://www.affymetrix.com).

Affymetrix used several genotyping quality control measures. To aid in sample tracking, sex-linked markers (X or Y chromosome) were genotyped on all samples to ensure that the DNA matched the expected sex for each individual. Further, positive and negative controls were run in parallel to ensure there was no contamination of the DNA. Other quality control measures included the addition of 25 CEPH family trios to the genotyping plates to test for non-Mendelian inheritance, and checking for high concordance of genotypes across sample pairs for potential unknown relationships, contamination, or mistracking. Samples that failed genotyping were defined by Affymetrix as those with a call rate (ie, the proportion of markers that gave unambiguous genotypes) less than 90% and repeatability rate (ie, sample-genotyped twice) less than 99%. Of the 1043 samples, 13 (1%) failed Affymetrix quality control criteria. Individual SNPs that failed genotyping were defined by Affymetrix as those with a call rate less than 80%, repeatability less than 99%, or non-Mendelian inheritance. A final number of 9237 SNPs were successfully genotyped out of 9412 attempted. Overall, the assay call rate was 99.13%, and the repeatability was 99.93%.

A second level of quality control was implemented at the Mayo Clinic. Of the 1030 samples that were successfully genotyped by Affymetrix, 48 were randomly selected duplicates (unknown to Affymetrix), leaving 982 unique subjects. We excluded from further analysis 1 man with heterozygous genotypes on the X chromosome, 11 nonwhite patients, 5 Hispanic patients, 22 patients with HL, and 1 patient later found to not have a lymphoma, leaving a final sample size of 942 genotyped subjects (458 patients and 484 controls) in the analysis.

Of the 9237 SNPs with genotype data provided by Affymetrix, we further excluded 524 with a call rate of less than 95%, 885 with a MAF of less than 1%, 3 with more than 2 genotype differences among the duplicates, and 5 with 1 or more male heterozygous genotypes for an X chromosome SNP. Furthermore, we excluded 85 SNPs that were not mapped in build 36 of the human genome (dbSNP; http://www.ncbi.nlm.nih.gov/projects/SNP/). This left 7735 SNPs that passed all quality controls.

Hardy-Weinberg equilibrium (HWE) was evaluated among the control subjects for each SNP using a chi-square test (MAF ≥ .05) or an exact test (MAF < .05). Because of multiple testing, SNPs found to be significant at a HWE threshold of .0001 or less were removed from further analyses. This threshold is conservative (ie, removing more SNPs than may be necessary); however, because not all SNPs were independent, a Bonferroni correction would be too liberal (ie, removing fewer SNPs than necessary). This removed 65 SNPs, and of these SNPs, we observed no clustering of HWE failures. All but 2 of these SNPs were independent of each other. In summary, we had 7670 SNPs (375 nsSNPs) for analysis.

Statistical analysis

The independent effects of the matching variables age (including its functional form), sex, and geographic region were examined in unconditional logistic regression models; geographic region was not significant (P > .2), and therefore was dropped from further consideration. We used unconditional logistic regression analysis to examine associations between each SNP and the risk of NHL, adjusting for the effects of age and sex. The most prevalent homozygous genotype was used as the reference group. Each polymorphism was modeled individually as having a log-additive effect in the regression model, and odds ratios (ORs) and 95% confidence intervals (CIs) were estimated. Associations between haplotypes from each gene and the risk of NHL were calculated using a score test implemented in HAPLO.SCORE16  from the Haplo.Stats S-plus library (http://mayoresearch.mayo.edu/mayo/research/schaid_lab/software.cfm). All SNPs located within a gene and SNPs located either 5 kb upstream or downstream were used in the haplotype analyses. Finally, we modeled the main effects for all independent (r2 < 0.25) SNPs from a gene in a multivariate logistic regression model. This approach does not require phase information and has been shown to have greater power than haplotype analysis.17  Our primary analysis approach for selecting noteworthy results was based on the gene-level tests, and genes that had a global multiple logistic regression or global haplotype P value of .001 or less were reported as noteworthy. Because nsSNPs are more likely have functional consequences, we separately selected noteworthy results for nsSNPs with a P value of .01 or less. In addition, we examined the overall significance of the P values for our gene-level and nsSNP-level tests using the tail strength methodology of Taylor and Tibshirani18 ; a QQ plot for gene-level P values of .10 or less is also provided as Figure S1 (available on the Blood website; see the Supplemental Materials link at the top of the online article). All analyses were done using S-plus (Insightful, Seattle, WA) or SAS (SAS Institute, Cary, NC).

To reduce the potential that population stratification affected our results, all analyses were restricted to subjects whose self-reported race was white. In addition, we tested our white subjects for potential population stratification by randomly selecting 1000 independent (r2 < 0.25) SNPs from our study and running the program Structure.19  We found no evidence for population structure in our data.

There were 458 patients with NHL and 484 controls in this analysis, and all were white. Patients and controls were well matched on age, sex, state of residence, and education level (Table 1). Patients were more likely to report a family history of NHL (4.8%) compared with controls (2.9%). The most common subtypes were CLL/small lymphocytic lymphoma (SLL) (N = 126), follicular lymphoma (N = 113), and diffuse large B-cell lymphoma (DLBCL) (N = 76).

Table 1

Characteristics of study participants

CharacteristicPatientsControls
No. 458 484 
Age, no. (%)   
    Younger than 40 y 32 (7.0) 29 (6.0) 
    40-49 y 78 (17.0) 54 (11.2) 
    50-59 y 83 (18.1) 103 (21.3) 
    60-69 y 143 (31.2) 139 (28.7) 
    70 y and older 122 (26.6) 159 (32.9) 
Age, mean y (± SD) 60.0 (± 13.6) 61.6 (± 12.9) 
Sex, no. (%)   
    Male 265 (57.9) 264 (54.5) 
    Female 193 (42.1) 220 (45.5) 
Residence, no. (%)   
    Minnesota 299 (65.3) 327 (67.6) 
    Iowa 90 (19.7) 88 (18.2) 
    Wisconsin 69 (15.1) 69 (14.3) 
Education level, no. (%)   
    Less than high school graduate 21 (5.9) 23 (5.4) 
    High school graduate/GED 93 (26.1) 93 (22.0) 
    Vocational/other postsecondary school 62 (17.4) 72 (17.0) 
    Some college/college graduate 129 (36.2) 149 (35.2) 
    Graduate or professional school 51 (14.3) 86 (20.3) 
    Missing 102 61 
Family history of NHL, no. (%)   
    No 339 (95.0) 406 (97.1) 
    Yes 18 (5.0) 12 (2.9) 
    Missing 101 66 
NHL subtype, no. (%)   
    SLL/CLL 126 (27.5) NA 
    Follicular 113 (24.7) NA 
    DLBCL 76 (16.6) NA 
    Marginal zone 31 (6.8) NA 
    Mantle cell 25 (5.5) NA 
    T-cell 17 (3.7) NA 
    Other/NOS 70 (15.3) NA 
CharacteristicPatientsControls
No. 458 484 
Age, no. (%)   
    Younger than 40 y 32 (7.0) 29 (6.0) 
    40-49 y 78 (17.0) 54 (11.2) 
    50-59 y 83 (18.1) 103 (21.3) 
    60-69 y 143 (31.2) 139 (28.7) 
    70 y and older 122 (26.6) 159 (32.9) 
Age, mean y (± SD) 60.0 (± 13.6) 61.6 (± 12.9) 
Sex, no. (%)   
    Male 265 (57.9) 264 (54.5) 
    Female 193 (42.1) 220 (45.5) 
Residence, no. (%)   
    Minnesota 299 (65.3) 327 (67.6) 
    Iowa 90 (19.7) 88 (18.2) 
    Wisconsin 69 (15.1) 69 (14.3) 
Education level, no. (%)   
    Less than high school graduate 21 (5.9) 23 (5.4) 
    High school graduate/GED 93 (26.1) 93 (22.0) 
    Vocational/other postsecondary school 62 (17.4) 72 (17.0) 
    Some college/college graduate 129 (36.2) 149 (35.2) 
    Graduate or professional school 51 (14.3) 86 (20.3) 
    Missing 102 61 
Family history of NHL, no. (%)   
    No 339 (95.0) 406 (97.1) 
    Yes 18 (5.0) 12 (2.9) 
    Missing 101 66 
NHL subtype, no. (%)   
    SLL/CLL 126 (27.5) NA 
    Follicular 113 (24.7) NA 
    DLBCL 76 (16.6) NA 
    Marginal zone 31 (6.8) NA 
    Mantle cell 25 (5.5) NA 
    T-cell 17 (3.7) NA 
    Other/NOS 70 (15.3) NA 

GED indicates general equivalency diploma; NA, not applicable; and NOS, not otherwise specified.

After all exclusions, there were 7670 SNPs available for analysis, and these SNPs were assigned to 1158 genes based on chromosomal position using build 36 of the human genome (dbSNP). The average number of SNPs per gene was 6.1, with a minimum of 1 SNP (in 318 genes) and a maximum of 136 SNPs for 1 gene. There were also a total of 375 nsSNPs available for analysis, and these were assigned to 262 genes. Table S1 reports all of the SNPs and their MAF among the controls.

Table 2 and 3 reports the gene level results based on the logistic regression and haplotype analyses, ranked by P value for all P values of .001 or less from either analysis. We also report results for the individual SNPs within these genes (Table 3). Based on the logistic regression analysis, the smallest P values were seen for CREB1 (P < .001) and FGG (P < .001). In CREB1, there were 5 SNPs with a P value of .05 or less, and the SNP with the smallest P value (.002) was rs2551919, with an ordinal OR of 0.69 (95% CI, 0.55-0.88). For FGG, there was only a single SNP (rs1800792), and the ordinal OR was 1.44 (95% CI, 1.19-1.73). The global P values from the haplotype results were similar to the logistic results. To assess the impact of multiple testing at the gene level, we calculated the tail strength of the 1158 P values from the logistic regression gene-based analysis. The tail strength was 0.10 (95% CI, 0.04-0.15), suggesting that our results identified 10% more signal than expected from chance.

Table 2

Logistic regression and haplotype results for genes with a P value of .001 or less

GeneLogistic regression results*
Haplotype results
SNPs 0.05 or less, no.Subtype results P
SNPs, no.PNo. SNPsNo. haplotypesGlobal PCLL/SLLFLDLBCL
CREB1 .0001 .001 .01 .02 .1 
FGG .0001 NA NA .0004 .001 .6 
MAP3K5 12 .0004 21 ND ND .05 .009 .2 
RIPK3 .0005 .001 .001 .3 .2 
LSP1 .0008 .003 .002 .03 .2 
TRAF1 .001 .01 .0001 .3 .4 
DUSP2 .001 NA NA .08 .2 .04 
ITGB3 .003 12 17 .001 .3 .02 .05 
GeneLogistic regression results*
Haplotype results
SNPs 0.05 or less, no.Subtype results P
SNPs, no.PNo. SNPsNo. haplotypesGlobal PCLL/SLLFLDLBCL
CREB1 .0001 .001 .01 .02 .1 
FGG .0001 NA NA .0004 .001 .6 
MAP3K5 12 .0004 21 ND ND .05 .009 .2 
RIPK3 .0005 .001 .001 .3 .2 
LSP1 .0008 .003 .002 .03 .2 
TRAF1 .001 .01 .0001 .3 .4 
DUSP2 .001 NA NA .08 .2 .04 
ITGB3 .003 12 17 .001 .3 .02 .05 

FL indicates follicular lymphoma; NA, not applicable; and ND, not done.

*

Number of SNPs used in the logistic regression model; P value from the global test.

Number of SNPs, haplotypes, and global P value from the haplotype model.

P value from logistic regression models for the NHL subtypes of CLL/SLL, FL, and DLBCL.

Gene name as defined in Entrez Gene.

Table 3

SNPs from genes with a P value of .001 or less from the logistic regression or haplotype analyses

Gene*, SNPSNP results
AllelesMAF patientsMAF controlsPOR (95% CI)SNPs in LR
CREB1       
    rs2551640 A/G 0.37 0.31 .009 1.30 (1.07, 1.59) — 
    rs2709359 G/A 0.021 0.037 .03 0.54 (0.30, 0.95) — 
    rs2709360 T/C 0.022 0.037 .05 0.57 (0.32, 1.00) × 
    rs2709402 C/A 0.17 0.22 .004 0.70 (0.56, 0.89) × 
    rs10932201 G/A 0.47 0.46 1.0 1.00 (0.83, 1.20) × 
    rs2551919 C/T 0.17 0.22 .002 0.69 (0.55, 0.88) — 
    rs3770704 T/C 0.016 0.009 .2 1.70 (0.73, 3.94) × 
FGG       
    rs1800792 A/G 0.51 0.42 .0002 1.44 (1.19, 1.73) × 
MAP3K5       
    rs2327742 A/G 0.19 0.14 .004 1.44 (1.12, 1.84) — 
    rs13203080 G/A 0.13 0.14 .2 0.85 (0.64, 1.11) — 
    rs7755097 T/C 0.18 0.14 .02 1.35 (1.05, 1.74) × 
    rs2327743 T/C 0.20 0.14 .002 1.48 (1.16, 1.89) — 
    rs2076262 T/C 0.02 0.00 .01 4.06 (1.34, 12.3) × 
    rs12164028 A/G 0.02 0.01 .02 3.08 (1.23, 7.71) × 
    rs2064205 C/T 0.38 0.38 .9 1.01 (0.84, 1.22) — 
    rs6906871 A/C 0.38 0.37 .7 1.03 (0.86, 1.24) — 
    rs2237269 G/C 0.16 0.17 .4 0.91 (0.71, 1.16) × 
    rs6904753 C/T 0.19 0.19 1.0 0.99 (0.79, 1.24) — 
    rs3765259 C/T 0.53 0.48 .08 1.17 (0.98, 1.39) × 
    rs9402839 G/A 0.10 0.11 .5 0.89 (0.65, 1.22) — 
    rs4363056 T/C 0.42 0.38 .09 1.18 (0.98, 1.42) — 
    rs7753357 T/A 0.07 0.09 .05 0.71 (0.51, 1.00) — 
    rs7748892 A/G 0.04 0.04 1.0 1.01 (0.63, 1.62) × 
    rs6570087 C/A 0.03 0.04 .3 0.74 (0.42, 1.27) × 
    rs10484491 A/G 0.49 0.46 .3 1.10 (0.91, 1.32) × 
    rs4896219 A/C 0.48 0.48 .9 1.02 (0.85, 1.22) × 
    rs932589 G/A 0.40 0.41 .6 0.95 (0.78, 1.15) × 
    rs9402845 G/A 0.38 0.39 .9 0.98 (0.82, 1.18) × 
    rs1570054 C/T 0.36 0.38 .4 0.91 (0.75, 1.11) × 
RIPK3       
    rs3212254 C/A 0.04 0.07 .004 0.55 (0.37, 0.82) × 
    rs724165 A/G 0.40 0.44 0.1 0.87 (0.72, 1.05) × 
    rs3212247 T/C 0.06 0.08 0.1 0.75 (0.52, 1.07) — 
    rs3212243 A/G 0.28 0.24 0.07 1.22 (0.99, 1.50) × 
LSP1       
    rs2089910 C/T 0.26 0.19 .0005 1.49 (1.19, 1.86) × 
    rs3817197 G/A 0.50 0.47 0.2 1.13 (0.93, 1.36) × 
TRAF1       
    rs4836834 A/T 0.45 0.41 .03 1.22 (1.02, 1.47) × 
    rs2269059 T/A 0.11 0.065 .0007 1.78 (1.28, 2.48) × 
    rs1930781 A/G 0.34 0.34 .8 1.02 (0.84, 1.24) — 
    rs3761846 T/C 0.45 0.40 .02 1.24 (1.03, 1.50) — 
    rs2416806 C/G 0.34 0.33 .8 1.03 (0.85, 1.25) — 
DUSP2       
    rs1724120 G/A 0.37 0.44 .001 0.74 (0.61, 0.89) × 
ITGB3       
    rs2317385 C/T 0.16 0.18 .6 0.93 (0.73, 1.19) — 
    rs2056131 C/T 0.34 0.30 .1 1.17 (0.96, 1.42) × 
    rs4525555 C/T 0.31 0.34 .1 0.87 (0.72, 1.05) — 
    rs10514919 G/T 0.23 0.29 .001 0.71 (0.58, 0.87) — 
    rs8073827 G/T 0.28 0.33 .03 0.80 (0.66, 0.97) — 
    rs3851806 G/C 0.17 0.18 .4 0.90 (0.70, 1.15) — 
    rs5918 T/C 0.13 0.18 .001 0.66 (0.52, 0.85) — 
    rs5919 T/C 0.08 0.05 .004 1.77 (1.20, 2.60) × 
    rs951351 C/T 0.04 0.04 .8 1.07 (0.67, 1.71) — 
    rs2292863 G/C 0.27 0.33 .002 0.73 (0.60, 0.90) — 
    rs3785872 G/A 0.10 0.11 .5 0.89 (0.66, 1.22) × 
    rs3809863 C/T 0.48 0.47 .5 1.06 (0.89, 1.27) × 
Gene*, SNPSNP results
AllelesMAF patientsMAF controlsPOR (95% CI)SNPs in LR
CREB1       
    rs2551640 A/G 0.37 0.31 .009 1.30 (1.07, 1.59) — 
    rs2709359 G/A 0.021 0.037 .03 0.54 (0.30, 0.95) — 
    rs2709360 T/C 0.022 0.037 .05 0.57 (0.32, 1.00) × 
    rs2709402 C/A 0.17 0.22 .004 0.70 (0.56, 0.89) × 
    rs10932201 G/A 0.47 0.46 1.0 1.00 (0.83, 1.20) × 
    rs2551919 C/T 0.17 0.22 .002 0.69 (0.55, 0.88) — 
    rs3770704 T/C 0.016 0.009 .2 1.70 (0.73, 3.94) × 
FGG       
    rs1800792 A/G 0.51 0.42 .0002 1.44 (1.19, 1.73) × 
MAP3K5       
    rs2327742 A/G 0.19 0.14 .004 1.44 (1.12, 1.84) — 
    rs13203080 G/A 0.13 0.14 .2 0.85 (0.64, 1.11) — 
    rs7755097 T/C 0.18 0.14 .02 1.35 (1.05, 1.74) × 
    rs2327743 T/C 0.20 0.14 .002 1.48 (1.16, 1.89) — 
    rs2076262 T/C 0.02 0.00 .01 4.06 (1.34, 12.3) × 
    rs12164028 A/G 0.02 0.01 .02 3.08 (1.23, 7.71) × 
    rs2064205 C/T 0.38 0.38 .9 1.01 (0.84, 1.22) — 
    rs6906871 A/C 0.38 0.37 .7 1.03 (0.86, 1.24) — 
    rs2237269 G/C 0.16 0.17 .4 0.91 (0.71, 1.16) × 
    rs6904753 C/T 0.19 0.19 1.0 0.99 (0.79, 1.24) — 
    rs3765259 C/T 0.53 0.48 .08 1.17 (0.98, 1.39) × 
    rs9402839 G/A 0.10 0.11 .5 0.89 (0.65, 1.22) — 
    rs4363056 T/C 0.42 0.38 .09 1.18 (0.98, 1.42) — 
    rs7753357 T/A 0.07 0.09 .05 0.71 (0.51, 1.00) — 
    rs7748892 A/G 0.04 0.04 1.0 1.01 (0.63, 1.62) × 
    rs6570087 C/A 0.03 0.04 .3 0.74 (0.42, 1.27) × 
    rs10484491 A/G 0.49 0.46 .3 1.10 (0.91, 1.32) × 
    rs4896219 A/C 0.48 0.48 .9 1.02 (0.85, 1.22) × 
    rs932589 G/A 0.40 0.41 .6 0.95 (0.78, 1.15) × 
    rs9402845 G/A 0.38 0.39 .9 0.98 (0.82, 1.18) × 
    rs1570054 C/T 0.36 0.38 .4 0.91 (0.75, 1.11) × 
RIPK3       
    rs3212254 C/A 0.04 0.07 .004 0.55 (0.37, 0.82) × 
    rs724165 A/G 0.40 0.44 0.1 0.87 (0.72, 1.05) × 
    rs3212247 T/C 0.06 0.08 0.1 0.75 (0.52, 1.07) — 
    rs3212243 A/G 0.28 0.24 0.07 1.22 (0.99, 1.50) × 
LSP1       
    rs2089910 C/T 0.26 0.19 .0005 1.49 (1.19, 1.86) × 
    rs3817197 G/A 0.50 0.47 0.2 1.13 (0.93, 1.36) × 
TRAF1       
    rs4836834 A/T 0.45 0.41 .03 1.22 (1.02, 1.47) × 
    rs2269059 T/A 0.11 0.065 .0007 1.78 (1.28, 2.48) × 
    rs1930781 A/G 0.34 0.34 .8 1.02 (0.84, 1.24) — 
    rs3761846 T/C 0.45 0.40 .02 1.24 (1.03, 1.50) — 
    rs2416806 C/G 0.34 0.33 .8 1.03 (0.85, 1.25) — 
DUSP2       
    rs1724120 G/A 0.37 0.44 .001 0.74 (0.61, 0.89) × 
ITGB3       
    rs2317385 C/T 0.16 0.18 .6 0.93 (0.73, 1.19) — 
    rs2056131 C/T 0.34 0.30 .1 1.17 (0.96, 1.42) × 
    rs4525555 C/T 0.31 0.34 .1 0.87 (0.72, 1.05) — 
    rs10514919 G/T 0.23 0.29 .001 0.71 (0.58, 0.87) — 
    rs8073827 G/T 0.28 0.33 .03 0.80 (0.66, 0.97) — 
    rs3851806 G/C 0.17 0.18 .4 0.90 (0.70, 1.15) — 
    rs5918 T/C 0.13 0.18 .001 0.66 (0.52, 0.85) — 
    rs5919 T/C 0.08 0.05 .004 1.77 (1.20, 2.60) × 
    rs951351 C/T 0.04 0.04 .8 1.07 (0.67, 1.71) — 
    rs2292863 G/C 0.27 0.33 .002 0.73 (0.60, 0.90) — 
    rs3785872 G/A 0.10 0.11 .5 0.89 (0.66, 1.22) × 
    rs3809863 C/T 0.48 0.47 .5 1.06 (0.89, 1.27) × 

LR indicates logistic regression; and —, not applicable.

*

Gene name as defined in Entrez Gene.

Ordinal OR, adjusted for age and sex.

SNPs (×) included in the logistic regression model.

Table 4 reports the results for the 6 nsSNPs ranked by the P value from the ordinal (log additive) model with a P value of .01 or less. The smallest P value was for ITGB3 (P = .001), and the ordinal OR was 0.66 (95% CI, 0.52-0.85). The tail strength for the nsSNP analysis (based on 375 P values) was 0.04 (−0.06-0.14), suggesting that none of these results were significant after accounting for multiple testing. Table 4 also reports the amino acid change for the nsSNPs; all of these changes were predicted to be benign using the software PolyPhen (Harvard University, Cambridge, MA; http://genetics.bwh.harvard.edu/pph).

Table 4

nsSNPs with a P value of .01 or less from the logistic regression analysis

SNPGeneNucleotide substitutionAmino acid changeResults for all NHL
Subtype results
MAF
PatientsControlsP, ordinalOR (95% CI)CLL/SLL OR (95% CI)FL OR (95% CI)DLBCL OR (95% CI)
rs5918 ITGB3 T/C L59P 0.13 0.18 .001 0.66 (0.52, 0.85) 0.82 (0.56, 1.19) 0.59 (0.38, 0.92) 0.31 (0.16, 0.61) 
rs5743815 TLR6 T/C V427A 0.02 0.004 .003 5.20 (1.77, 15.3) 8.74 (2.51, 30.4) 4.59 (1.10, 19.2) 4.30 (0.92, 20.2) 
rs7300972 SELPLG T/C M264V 0.03 0.01 .003 3.20 (1.48, 6.91) 2.03 (0.66, 6.23) 3.02 (1.05, 8.68) 2.94 (0.87, 9.85) 
rs2072797 UNC84B C/T G671S 0.14 0.10 .006 1.50 (1.12, 2.00) 1.66 (1.11, 2.48) 1.24 (0.77, 1.97) 1.50 (0.90, 2.51) 
rs36686 B3GNT3 A/G H328R 0.21 0.26 .009 0.74 (0.59, 0.93) 0.52 (0.35, 0.76) 1.02 (0.72, 1.43) 0.89 (0.59, 1.34) 
rs3132453 BAT2 C/A V1883L 0.06 0.09 .01 0.64 (0.45, 0.90) 0.56 (0.31, 1.01) 0.67 (0.38, 1.19) 0.56 (0.27, 1.16) 
SNPGeneNucleotide substitutionAmino acid changeResults for all NHL
Subtype results
MAF
PatientsControlsP, ordinalOR (95% CI)CLL/SLL OR (95% CI)FL OR (95% CI)DLBCL OR (95% CI)
rs5918 ITGB3 T/C L59P 0.13 0.18 .001 0.66 (0.52, 0.85) 0.82 (0.56, 1.19) 0.59 (0.38, 0.92) 0.31 (0.16, 0.61) 
rs5743815 TLR6 T/C V427A 0.02 0.004 .003 5.20 (1.77, 15.3) 8.74 (2.51, 30.4) 4.59 (1.10, 19.2) 4.30 (0.92, 20.2) 
rs7300972 SELPLG T/C M264V 0.03 0.01 .003 3.20 (1.48, 6.91) 2.03 (0.66, 6.23) 3.02 (1.05, 8.68) 2.94 (0.87, 9.85) 
rs2072797 UNC84B C/T G671S 0.14 0.10 .006 1.50 (1.12, 2.00) 1.66 (1.11, 2.48) 1.24 (0.77, 1.97) 1.50 (0.90, 2.51) 
rs36686 B3GNT3 A/G H328R 0.21 0.26 .009 0.74 (0.59, 0.93) 0.52 (0.35, 0.76) 1.02 (0.72, 1.43) 0.89 (0.59, 1.34) 
rs3132453 BAT2 C/A V1883L 0.06 0.09 .01 0.64 (0.45, 0.90) 0.56 (0.31, 1.01) 0.67 (0.38, 1.19) 0.56 (0.27, 1.16) 

Ordinal OR and 95% CIs were adjusted for age and sex.

As a secondary analysis, we evaluated the gene level (Table 2; Table S2) and nsSNP (Table 4) associations for the 3 most common NHL subtypes in our dataset. These analyses have much less power (due to smaller sample size) and have not been corrected for multiple testing (a nominal P ≤ .05 was used for this analysis), and therefore should be interpreted with caution. With these caveats in mind, several potentially interesting patterns emerged. For CLL/SLL (N = 126 patients), associations at the gene level (from logistic regression models) were statistically significant at P values of .05 or less for most genes found to be notable in the main analysis; the exceptions included DUSP2 (P = .08) and ITGB3 (P = .3). For the nsSNPs, all ORs for CLL/SLL were of a similar magnitude as those for all NHL, although only nsSNPs for TLR6, UNC84B, and B3GNT3 were statistically significant at P values of .05 or less. For follicular lymphoma (N = 113 patients), FGG (P = .001), MAP3K5 (P = .009), CREB1 (P = .02), ITGB3 (P = .02), and LSP1 (P = .03) were associated with risk at the gene level. In nsSNP analysis, with the exception of B3GNT3, all ORs were in the same direction and of similar magnitude as the ORs for all NHL, although only nsSNPs in ITGB3, TLR6, and SELPLG achieved a P value of .05 or less. For DLBCL (N = 76 patients), DUSP2 (P = .04) and ITGB3 (P = .05) were associated with risk at the gene level. In the nsSNP analysis, all ORs were in the same direction and of similar magnitude as for all NHL, although only the association for the nsSNP in ITGB3 was statistically significant at P values of .05 or less.

In a discovery exercise, we used a panel of genes related to immune function and inflammation to identify genetic risk factors for NHL. After exclusion of SNPs that did not meet our strict quality control or that had a MAF of less than 0.01, we had data available for 7670 SNPs assigned to 1158 genes. We identified 9 genes and 6 nsSNPs (from 6 genes) of greatest interest based on our statistical criteria. These types of analyses are prone to false discovery due to the large number of statistical tests that are conducted, and while the signal from our gene-level analysis suggested that we observed a 10% greater signal over that of chance, our nsSNP findings were consistent with a chance finding after accounting for the number of tests conducted. Ultimately, replication of genetic associations in independent studies or within a consortium such as InterLymph8  is required, and the genes we identified are excellent candidates.

This is the largest evaluation of genes involved in immunity and inflammation reported to date for NHL. While our findings must be considered preliminary and hypothesis-generating, we observed several patterns among the highest-ranked genes that are supported by the limited existing literature on genetic susceptibility to NHL (Table 5). The strongest findings to date have been for a role for specific SNPs from genes involved in inflammation and immune response, particularly TNF and LTA,7,9,59  although smaller studies have not observed these associations,60,61  perhaps due to lower power. While these specific SNPs from TNF and LTA were not on our SNP panel, we did identify several genes important in the inflammatory and innate immune response, including TRAF, RIPK3, BAT2, and TLR6. TRAF1 expression has been shown to be elevated in NHL52  and particularly CLL,52,53  and in subtype analysis, CLL/SLL was most strongly associated with TRAF1. RIPK3 is a component of the TNFR1 signaling complex, and BAT2 is located in the HLA class III complex in the vicinity of TNF and LTA and has been associated with autoimmune disease.23,24  Even though the nsSNP for BAT2 (V1883L) is predicted to be benign using PolyPhen, further analysis in this gene-rich area is warranted. Toll-like receptors (TLRs) bridge the innate and adaptive immune systems,62  and the nsSNP in TLR6 (rs5743815) that was associated with risk in this study has not previously been evaluated in NHL. Little is known about the functional consequences, if any, of this SNP, although the substitution (V427A) was predicted to be benign using PolyPhen. Although the MAF was low in patients (0.02) and controls (0.004), the association was strong for NHL overall (OR = 5.20; 95% CI, 1.77-15.3) and for each of the subtypes. A nsSNP from TLR2 (−16933T > A) has been reported to be associated with NHL overall and with follicular lymphoma in particular,63  and a nsSNP in TLR4 (rs4986790; 1063A > G) was associated with DLBCL in 1 study,12  although this was not observed in 2 other studies.9,63 

Table 5

Summary of top-ranked genes

Gene (aliases)*ChromosomeGene ID no.*Gene description*Gene Ontology (GO)CommentsSelected references
B3GNT3 (TMEM319p13.1 10331 UDP-GlcNAc:betaGal beta-1,3-N-acetylglucos-aminyltransferase 3 Protein amino acid glycosylation B3GNT3 encodes for beta-1,3-N-acetylglucosaminyltransferase 3, an enzyme that is classified as a type II transmembrane protein. B3GNT3 is involved in the biosynthesis of poly-N-acetyllactosamine chains and the biosynthesis of the backbone structure of dimeric sialyl Lewis a. B3GNT3 also plays dominant roles in L-selectin ligand biosynthesis, which is important for lymphocyte homing and trafficking. 20,22  
BAT2 6p21.3 7916 HLA-B associated transcript 2 Protein binding Part of a cluster of genes (BAT1-BAT5) in the HLA complex class III region (in vicinity of TNF and LTA). Gene has been associated with insulin-dependent diabetes and rheumatoid arthritis risk and response to therapy. 23,25  
CREB1 2q32.3–34 1385 cAMP responsive element binding protein 1 Regulation of transcription; signal transduction; regulation of cell size Transcription factor that participates in a variety of cellular processes, including growth factor-dependent survival and proliferation, as well as glucose homeostasis. Proto-oncogene for clear cell soft tissue sarcoma, AML, T-cell leukemias, follicular and transformed NHL, and myeloproliferative disease. CREB binds CRE site in the promoter of translocated bcl-2 in FL with t14;18 translocations. 26,,,30  
DUSP2 (PAC12q11 1844 Dual specificity phosphatase 2 Inactivation of MAPK activity; protein AA dephosphorylation DUSP2 is a member of the dual specificity protein phosphatase subfamily, which negatively regulates members of the mitogen-activated protein (MAP) kinase superfamily (MAPK/ERK, SAPK/JNK, p38). DUSP2 is predominantly expressed in hematopoietic lineages. Reduction of DUSP2 transcription using small interfering RNA inhibits p53-mediated apoptosis, whereas overexpression of DUSP2 increases susceptibility to apoptosis and suppresses tumor formation. DUSP2 knock out mice show reduced cytokine production and are protected from inflammatory arthritis. 31,,,35  
FGG 4q28 2266 Fibrinogen gamma chain Blood pressure regulation; platelet activation; positive regulation of cell proliferation; protein polymerization; signal transduction FGG encodes gamma component of fibrinogen, a blood-borne glycoprotein comprised of three pairs of non-identical polypeptide chains. Important role in tissue repair by providing an initial matrix that can stabilize wound fields and support local cell proliferation and migration. Mutations in this gene lead to several disorders, including dysfibrinogenemia, hypofibrinogenemia and thrombophilia; polymorphisms have been associated with serum fibrinogen levels. 36,38  
ITGB3 (CD6117q21.32 3690 Integrin, beta 3 (platelet glycoprotein IIIa, antigen CD61) Blood coagulation; cell adhesion; cell-matrix adhesion; development; integrin-mediated signaling pathway Integrins are cell-surface proteins composed of an alpha and beta chain; they participate in cell adhesion and cell-surface mediated signaling. ITGB3 codes for glycoprotein IIIa (GPIIIa), and along with alpha IIb chain forms the platelet adhesive protein receptor complex glycoprotein IIb/IIIa. It is also the beta subunit found in the fibronectin and vitronectin receptors. ITGB3 rs5918 results in a substitution (Leu33Pro) which has been associated with risk of ovarian and breast cancer and melanoma. 39  
LSP1 11p15.5 4046 Lymphocyte-specific protein 1 Cell motility; cellular defense response; signal transduction LSP1 encodes an intracellular F-actin binding protein, which is expressed in lymphocytes, neutrophils, macrophages and endothelium. Appears to regulate neutrophil motility, adhesion to fibrinogen matrix proteins, and transendothelial migration. 40  
MAP3K5 (ASK16q22.33 4217 MAPK kinase kinase MAPK kinase kinase cascade; activation of JNK activity; apoptosis; response to stress MAPKKK5 contains 1,374 amino acids with all 11 kinase subdomains. MAP3K5 is an upstream activator of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase signaling cascades. Overexpression of ASK1 induced apoptotic cell death, and ASK1 was activated in cells treated with TNF-α. 41,43  
RIPK3 (RIP314q11.2 11035 Receptor-interacting serine-threonine kinase 3 Apoptosis; protein amino acid phosphorylation; signal transduction Gene product is a member of the receptor-interacting protein (RIP) family of serine/threonine protein kinases. RIPK3 is a component of the TNFR1 signaling complex, and can induce apoptosis and weakly activate the NF-κB transcription factor. A splice variant is increased in colon and lung tumors relative to matched normals. 44,46  
SELPLG (CD162, PSGL112q24 6404 Selectin P ligand Cell adhesion Selectin P ligand (also know as P-selectin glycoprotein ligand) is the high affinity counter-receptor for P-selectin on myeloid cells and stimulated T lymphocytes. Plays a critical role in the tethering of these cells to activated platelets or endothelial cells expressing P-selectin. PSGL-1 enhances tyrosine phosphorylation, activates ERK-1 and ERK-2 through MEK (MAP kinase kinase), and stimulates IL-8 secretion in neutrophils. 4748  
TLR6 4p14 10333 Toll-like receptor 6 T-helper 1 type immune response; activation of NF-kappaB-inducing kinase; defense response to bacterium; innate immune response; macrophage activation Member of the IL-1R superfamily; TLR6 is a member of the TLR2 subfamily, and forms a heterodimer with TLR2. TLRs recognize conserved motifs of microbial origin (pathogen-associated molecular patterns, PAMPs). Activation of TLR2/6 results in secretion of pro-inflammatory cytokines. TLR6 expression has been demonstrated in mast cells, monocytes, and platelets. 49,  
TRAF1 9q33-q34 7185 TNF receptor-associated factor 1 Protein complex assembly; regulation of apoptosis; signal transduction Member of the TNF receptor (TNFR) associated factor (TRAF) protein family. TRAF1 and TRAF2 form a heterodimeric complex, which is required for TNF-alpha-mediated activation of MAPK8/JNK and NF-kappaB; this complex also mediates the anti-apoptotic signals from TNF receptors. Elevated TRAF1 expression in NHL and CLL. 52,,,56  
UNC84B (KIAA0668, SUN222q13.1 25777 unc-84 homolog B (C elegansMitotic spindle organization and biogenesis; nuclear migration UNC84B, also known as SUN2, codes for a mammalian inner nuclear membrane protein. Sun1 and Sun2 may form a physical interaction between the nuclear envelope and centrosome. 5758  
Gene (aliases)*ChromosomeGene ID no.*Gene description*Gene Ontology (GO)CommentsSelected references
B3GNT3 (TMEM319p13.1 10331 UDP-GlcNAc:betaGal beta-1,3-N-acetylglucos-aminyltransferase 3 Protein amino acid glycosylation B3GNT3 encodes for beta-1,3-N-acetylglucosaminyltransferase 3, an enzyme that is classified as a type II transmembrane protein. B3GNT3 is involved in the biosynthesis of poly-N-acetyllactosamine chains and the biosynthesis of the backbone structure of dimeric sialyl Lewis a. B3GNT3 also plays dominant roles in L-selectin ligand biosynthesis, which is important for lymphocyte homing and trafficking. 20,22  
BAT2 6p21.3 7916 HLA-B associated transcript 2 Protein binding Part of a cluster of genes (BAT1-BAT5) in the HLA complex class III region (in vicinity of TNF and LTA). Gene has been associated with insulin-dependent diabetes and rheumatoid arthritis risk and response to therapy. 23,25  
CREB1 2q32.3–34 1385 cAMP responsive element binding protein 1 Regulation of transcription; signal transduction; regulation of cell size Transcription factor that participates in a variety of cellular processes, including growth factor-dependent survival and proliferation, as well as glucose homeostasis. Proto-oncogene for clear cell soft tissue sarcoma, AML, T-cell leukemias, follicular and transformed NHL, and myeloproliferative disease. CREB binds CRE site in the promoter of translocated bcl-2 in FL with t14;18 translocations. 26,,,30  
DUSP2 (PAC12q11 1844 Dual specificity phosphatase 2 Inactivation of MAPK activity; protein AA dephosphorylation DUSP2 is a member of the dual specificity protein phosphatase subfamily, which negatively regulates members of the mitogen-activated protein (MAP) kinase superfamily (MAPK/ERK, SAPK/JNK, p38). DUSP2 is predominantly expressed in hematopoietic lineages. Reduction of DUSP2 transcription using small interfering RNA inhibits p53-mediated apoptosis, whereas overexpression of DUSP2 increases susceptibility to apoptosis and suppresses tumor formation. DUSP2 knock out mice show reduced cytokine production and are protected from inflammatory arthritis. 31,,,35  
FGG 4q28 2266 Fibrinogen gamma chain Blood pressure regulation; platelet activation; positive regulation of cell proliferation; protein polymerization; signal transduction FGG encodes gamma component of fibrinogen, a blood-borne glycoprotein comprised of three pairs of non-identical polypeptide chains. Important role in tissue repair by providing an initial matrix that can stabilize wound fields and support local cell proliferation and migration. Mutations in this gene lead to several disorders, including dysfibrinogenemia, hypofibrinogenemia and thrombophilia; polymorphisms have been associated with serum fibrinogen levels. 36,38  
ITGB3 (CD6117q21.32 3690 Integrin, beta 3 (platelet glycoprotein IIIa, antigen CD61) Blood coagulation; cell adhesion; cell-matrix adhesion; development; integrin-mediated signaling pathway Integrins are cell-surface proteins composed of an alpha and beta chain; they participate in cell adhesion and cell-surface mediated signaling. ITGB3 codes for glycoprotein IIIa (GPIIIa), and along with alpha IIb chain forms the platelet adhesive protein receptor complex glycoprotein IIb/IIIa. It is also the beta subunit found in the fibronectin and vitronectin receptors. ITGB3 rs5918 results in a substitution (Leu33Pro) which has been associated with risk of ovarian and breast cancer and melanoma. 39  
LSP1 11p15.5 4046 Lymphocyte-specific protein 1 Cell motility; cellular defense response; signal transduction LSP1 encodes an intracellular F-actin binding protein, which is expressed in lymphocytes, neutrophils, macrophages and endothelium. Appears to regulate neutrophil motility, adhesion to fibrinogen matrix proteins, and transendothelial migration. 40  
MAP3K5 (ASK16q22.33 4217 MAPK kinase kinase MAPK kinase kinase cascade; activation of JNK activity; apoptosis; response to stress MAPKKK5 contains 1,374 amino acids with all 11 kinase subdomains. MAP3K5 is an upstream activator of c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase signaling cascades. Overexpression of ASK1 induced apoptotic cell death, and ASK1 was activated in cells treated with TNF-α. 41,43  
RIPK3 (RIP314q11.2 11035 Receptor-interacting serine-threonine kinase 3 Apoptosis; protein amino acid phosphorylation; signal transduction Gene product is a member of the receptor-interacting protein (RIP) family of serine/threonine protein kinases. RIPK3 is a component of the TNFR1 signaling complex, and can induce apoptosis and weakly activate the NF-κB transcription factor. A splice variant is increased in colon and lung tumors relative to matched normals. 44,46  
SELPLG (CD162, PSGL112q24 6404 Selectin P ligand Cell adhesion Selectin P ligand (also know as P-selectin glycoprotein ligand) is the high affinity counter-receptor for P-selectin on myeloid cells and stimulated T lymphocytes. Plays a critical role in the tethering of these cells to activated platelets or endothelial cells expressing P-selectin. PSGL-1 enhances tyrosine phosphorylation, activates ERK-1 and ERK-2 through MEK (MAP kinase kinase), and stimulates IL-8 secretion in neutrophils. 4748  
TLR6 4p14 10333 Toll-like receptor 6 T-helper 1 type immune response; activation of NF-kappaB-inducing kinase; defense response to bacterium; innate immune response; macrophage activation Member of the IL-1R superfamily; TLR6 is a member of the TLR2 subfamily, and forms a heterodimer with TLR2. TLRs recognize conserved motifs of microbial origin (pathogen-associated molecular patterns, PAMPs). Activation of TLR2/6 results in secretion of pro-inflammatory cytokines. TLR6 expression has been demonstrated in mast cells, monocytes, and platelets. 49,  
TRAF1 9q33-q34 7185 TNF receptor-associated factor 1 Protein complex assembly; regulation of apoptosis; signal transduction Member of the TNF receptor (TNFR) associated factor (TRAF) protein family. TRAF1 and TRAF2 form a heterodimeric complex, which is required for TNF-alpha-mediated activation of MAPK8/JNK and NF-kappaB; this complex also mediates the anti-apoptotic signals from TNF receptors. Elevated TRAF1 expression in NHL and CLL. 52,,,56  
UNC84B (KIAA0668, SUN222q13.1 25777 unc-84 homolog B (C elegansMitotic spindle organization and biogenesis; nuclear migration UNC84B, also known as SUN2, codes for a mammalian inner nuclear membrane protein. Sun1 and Sun2 may form a physical interaction between the nuclear envelope and centrosome. 5758  
*

As defined in Entrez Gene.

Our data also suggest a role for genetic variation in signal transduction pathways, particularly related to TNF and TLR signaling. MAP3K5 belongs to the mitogen-activated protein kinase (MAPK) pathway, and it can be activated by TNFα through interactions with TNF receptor 1 (TNFR1) and TNFR2, TNFR-associated factors (TRAFs), and TNFR-associated death domain (TRADD).41,42 DUSP2 is highly inducible and encodes a protein that is predominately expressed in hematopoeitic lineages, particularly immune cells infiltrating inflammatory lesions.64  DUSP2 primarily inactivates p38 MAPK and ERK1 and ERK2, and thus can regulate transcription factors like CREB and AP-1.31 CREB1 encodes a transcription factor that mediates response to a variety of growth and stress signals,26  and is a putative oncogene that has been implicated in several cancers, including myeloid neoplasia and follicular and transformed NHL.27  CREB is activated by TNF/TNFR1 signaling through a p38MAPK/MSK1 signaling pathway and through TLR signaling.65,66  Of note, the associations for MAP3K5, DUSP2, and CREB1 were not specific to any NHL subtype, suggesting these genes may play a more global role in lymphomagenesis.

Genes involved in lymphocyte trafficking and migration were also in our top hits. B3GNT3 plays an important role in L-selectin biosynthesis,20  and SELPLG encodes a ligand for P-selectin (PSGL-1), which is important for tethering and rolling of leukocytes to endothelial cells and platelets67  and also may activate β2 integrins in neutrophils.68  Selectins appear to play an important role in tumor cell survival and metastasis,69  and increased soluble P-selectin has been reported in breast and hematologic cancers.70 

Our final and perhaps most unexpected observation was that genes involved in the acute-phase response and the coagulation-fibrinolytic system, specifically FGG and ITGB3, were associated with NHL risk. There is accumulating experimental evidence for a role of fibrin and fibrinogen degradation products in carcinogenesis by promoting invasive growth, angiogenesis, and metastasis.36,71,72 ITGB3 codes for the beta subunit of the glycoprotein IIb/IIIa complex, which mediates platelet aggregation by serving as a receptor for fibrinogen. The ITGB3 nsSNP rs5918 results in a substitution (Leu59Pro, also reported as Leu33Pro) that introduces a nick in the polypeptide chain, and this SNP leads to increased binding of fibrinogen to the IIb/IlIa complex,73  increased platelet aggregation,74  decreased bleeding time,75  and increased signaling through ERK2 of the MAPK pathway.76  In contrast to our results, this SNP has been associated with risk of all cancer in a Danish cohort study (relative risk [RR] for proline/proline genotype was 1.4; 95% CI 1.1-1.9).39  In subsite analysis of the Danish cohort, risks were specific to ovarian and breast cancer and melanoma; results specific to NHL were not reported (there were only 26 patients). Patients with NHL are at increased risks for coagulation disorders,36  and patients with venous thromboembolism have been found to have increased risk of NHL even more than 2 years after admission (standardized incidence ratio [SIR] = 1.4; 95% CI 1.2-1.6).77 

Strengths of this study include the careful quality control in genotyping and the use of the HapMap to tag the genes of interest, as well as inclusion of nsSNPs from these genes. Although our panel had 9412 SNPs, only 7670 (81%) were available for analysis, and this decreased the average number of SNPs from 6.8 to 6.1 per gene. In terms of SNP coverage against the current version of the HapMap, defined as the number of SNPs in a gene with a MAF of 0.05 or more for which we had a tagSNP (based on r2 = 0.8) divided by the total number of SNPs with a MAF of 0.05 or more, the median coverage was 61.0% (range, 0.3%-100%). Thus, our study may have missed important genes in the etiology of NHL due to low coverage of many of the genes. The most common reasons for exclusion of an SNP included SNPs with a MAF of less than 1% (N = 885 of 1742 SNPs; 51%) and SNPs with a call rate of less than 95% (N = 524 of 1742 SNPs; 30%). The large number of exclusions for low MAF was mainly due to the inclusion of SNPs from other ethnic populations in the SNP panel, which had little or no variation in our white population. Based on the restriction of our analysis to whites and the use of the Structure program, our results are not likely to be confounded by population stratification. While this restriction increases the internal validity of the results, it does decrease the generalizability of the findings to other racial/ethnic groups. Our study was not population-based, but we have carefully designed our clinic-based study to adhere to basic epidemiologic principles of the case-control study design, and in particular our controls are derived from the same underlying population source that generated our patients and were not selected on basis of any particular medical history or other exposure (including genotype). We also restricted case ascertainment to the 3-state region surrounding Mayo Clinic Rochester in order to reduce referral bias that can occur with patients coming from farther distances. Of note, hospital-based and population-based studies have reported similar allele and genotype frequencies among controls for a variety of metabolic genes.78 

In summary, our study provides additional evidence of the important role played by genes involved in the immune response in the etiology of NHL, particularly with respect to TNF and TLR pathways and MAPK signaling. Our study further suggests genes involved in lymphocyte trafficking and migration and coagulation pathways may also play important roles. Beyond the need to replicate these genes, additional work will need to be conducted to identify the causative SNPs in the candidate genes (eg, by fine mapping), the role of genes up and downstream of the candidate gene(s), further evaluation within NHL subtypes with sufficient sample size, and ultimately interactions with environmental exposures. Such approaches should help identify new etiologic pathways and high-risk populations for NHL, and this information should ultimately aid in identifying preventive strategies for this malignancy.

The online version of this article contains a data supplement.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

We thank Sondra Buehler for her editorial assistance.

This work was supported by National Institutes of Health grants R01 CA92153 (J.R.C.) and K07 CA94919 (S.L.S.).

National Institutes of Health

Contribution: J.R.C. designed the study, obtained funding, and drafted the manuscript; S.M.A., T.G.C., A.D., T.M.H., D.J., N.E.K., M.L., W.R.M., and T.E.W. gathered clinical, laboratory, and pathology data; J.M.C. and W.L.-M. performed bioinformatics; Z.S.F., A.H.W., and S.L.S. (with input from J.R.C.) performed statistical analysis; and all authors revised the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: James R. Cerhan, Department of Health Sciences Research, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905; e-mail:cerhan.james@mayo.edu.

1
Morton
LM
Wang
SS
Devesa
SS
Hartge
P
Weisenburger
DD
Linet
MS
Lymphoma incidence patterns by WHO subtype in the United States, 1992–2001.
Blood
2006
107
265
276
2
Jemal
A
Siegel
R
Ward
E
Murray
T
Xu
J
Thun
MJ
Cancer statistics, 2007.
CA Cancer J Clin
2007
57
43
66
3
Alexander
DD
Mink
PJ
Adami
HO
et al
The non-Hodgkin lymphomas: a review of the epidemiologic literature.
Int J Cancer
2007
120
1
39
4
Goldgar
DE
Easton
DF
Cannon-Albright
LA
Skolnick
MH
Systematic population-based assessment of cancer risk in first-degree relatives of cancer probands.
J Natl Cancer Inst
1994
86
1600
1608
5
Wang
SS
Slager
SL
Brennan
P
et al
Family history of hematopoietic malignancies and risk of non-Hodgkin lymphoma (NHL): a pooled analysis of 10 211 cases and 11 905 controls from the International Lymphoma Epidemiology Consortium (InterLymph).
Blood
2007
109
3479
3488
6
Collins
FS
Guyer
MS
Charkravarti
A
Variations on a theme: cataloging human DNA sequence variation.
Science
1997
278
1580
1581
7
Fitzgibbon
J
Grenzelias
D
Matthews
J
Lister
TA
Gupta
RK
Tumour necrosis factor polymorphisms and susceptibility to follicular lymphoma.
Br J Haematol
1999
107
388
391
8
Rothman
N
Skibola
CF
Wang
SS
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
9
Wang
SS
Cerhan
JR
Hartge
P
et al
Common genetic variants in proinflammatory and other immunoregulatory genes and risk for non-hodgkin lymphoma.
Cancer Res
2006
66
9771
9780
10
Lech-Maranda
E
Baseggio
L
Bienvenu
J
et al
Interleukin-10 gene promoter polymorphisms influence the clinical outcome of diffuse large B-cell lymphoma.
Blood
2004
103
3529
3534
11
Lan
Q
Zheng
T
Rothman
N
et al
Cytokine polymorphisms in the Th1/Th2 pathway and susceptibility to non-Hodgkin lymphoma.
Blood
2006
107
4101
4108
12
Forrest
MS
Skibola
CF
Lightfoot
TJ
et al
Polymorphisms in innate immunity genes and risk of non-Hodgkin lymphoma.
Br J Haematol
2006
134
180
183
13
Jaffe
ES
Harris
N
Stein
H
Vardiman
J
World Health Organization classification of tumours pathology and genetics, tumours of hematopoietic and lymphoid tissues.
2001
Lyon, France
IARC Press
14
Hardenbol
P
Yu
F
Belmont
J
et al
Highly multiplexed molecular inversion probe genotyping: over 10,000 targeted SNPs genotyped in a single tube assay.
Genome Res
2005
15
269
275
15
Moorhead
M
Hardenbol
P
Siddiqui
F
et al
Optimal genotype determination in highly multiplexed SNP data.
Eur J Hum Genet
2006
14
207
215
16
Schaid
DJ
Rowland
CM
Tines
DE
Jacobson
RM
Poland
GA
Score tests for association between traits and haplotypes when linkage phase is ambiguous.
Am J Hum Genet
2002
70
425
434
17
Chapman
JM
Cooper
JD
Todd
JA
Clayton
DG
Detecting disease associations due to linkage disequilibrium using haplotype tags: a class of tests and the determinants of statistical power.
Hum Hered
2003
56
18
31
18
Taylor
J
Tibshirani
R
A tail strength measure for assessing the overall univariate significance in a dataset.
Biostatistics
2006
7
167
181
19
Pritchard
JK
Stephens
M
Donnelly
P
Inference of population structure using multilocus genotype data.
Genetics
2000
155
945
959
20
Shiraishi
N
Natsume
A
Togayachi
A
et al
Identification and characterization of three novel beta-1,3-N-acetylglucosaminyltransferases structurally related to the beta-1,3-galactosyltransferase family.
J Biol Chem
2001
276
3498
3507
21
Hakomori
S
Tumor malignancy defined by aberrant glycosylation and sphingo(glyco)lipid metabolism.
Cancer Res
1996
56
5309
5318
22
Kobata
A
Amano
J
Altered glycosylation of proteins produced by malignant cells, and application for the diagnosis and immunotherapy of tumours.
Immunol Cell Biol
2005
83
429
439
23
Hashimoto
M
Nakamura
N
Obayashi
H
et al
Genetic contribution of the BAT2 gene microsatellite polymorphism to the age-at-onset of insulin-dependent diabetes mellitus.
Hum Genet
1999
105
197
199
24
Singal
DP
Li
J
Zhu
Y
HLA class III region and susceptibility to rheumatoid arthritis.
Clin Exp Rheumatol
2000
18
485
491
25
Martinez
A
Salido
M
Bonilla
G
et al
Association of the major histocompatibility complex with response to infliximab therapy in rheumatoid arthritis patients.
Arthritis Rheum
2004
50
1077
1082
26
Kinjo
K
Sandoval
S
Sakamoto
KM
Shankar
DB
The role of CREB as a proto-oncogene in hematopoiesis.
Cell Cycle
2005
4
1134
1135
27
Arcinas
M
Heckman
CA
Mehew
JW
Boxer
LM
Molecular mechanisms of transcriptional control of bcl-2 and c-myc in follicular and transformed lymphoma.
Cancer Res
2001
61
5202
5206
28
Ji
L
Mochon
E
Arcinas
M
Boxer
LM
CREB proteins function as positive regulators of the translocated bcl-2 allele in t(14;18) lymphomas.
J Biol Chem
1996
271
22687
22691
29
Conkright
MD
Montminy
M
CREB: the unindicted cancer co-conspirator.
Trends Cell Biol
2005
15
457
459
30
Shankar
DB
Cheng
JC
Kinjo
K
et al
The role of CREB as a proto-oncogene in hematopoiesis and in acute myeloid leukemia.
Cancer Cell
2005
7
351
362
31
Lang
R
Hammer
M
Mages
J
DUSP meet immunology: dual specificity MAPK phosphatases in control of the inflammatory response.
J Immunol
2006
177
7497
7504
32
Rohan
PJ
Davis
P
Moskaluk
CA
et al
PAC-1: a mitogen-induced nuclear protein tyrosine phosphatase.
Science
1993
259
1763
1766
33
Ward
Y
Gupta
S
Jensen
P
Wartmann
M
Davis
RJ
Kelly
K
Control of MAP kinase activation by the mitogen-induced threonine/tyrosine phosphatase PAC1.
Nature
1994
367
651
654
34
Yin
Y
Liu
YX
Jin
YJ
Hall
EJ
Barrett
JC
PAC1 phosphatase is a transcription target of p53 in signalling apoptosis and growth suppression.
Nature
2003
422
527
531
35
Jeffrey
KL
Brummer
T
Rolph
MS
et al
Positive regulation of immune cell function and inflammatory responses by phosphatase PAC-1.
Nat Immunol
2006
7
274
283
36
Boccaccio
C
Medico
E
Cancer and blood coagulation.
Cell Mol Life Sci
2006
63
1024
1027
37
Scott
EM
Ariens
RA
Grant
PJ
Genetic and environmental determinants of fibrin structure and function: relevance to clinical disease.
Arterioscler Thromb Vasc Biol
2004
24
1558
1566
38
Uitte de Willige
S
de Visser
MC
Houwing-Duistermaat
JJ
Rosendaal
FR
Vos
HL
Bertina
RM
Genetic variation in the fibrinogen gamma gene increases the risk for deep venous thrombosis by reducing plasma fibrinogen gamma levels.
Blood
2005
106
4176
4183
39
Bojesen
SE
Tybjaerg-Hansen
A
Nordestgaard
BG
Integrin beta3 Leu33Pro homozygosity and risk of cancer.
J Natl Cancer Inst
2003
95
1150
1157
40
May
W
Korenberg
JR
Chen
XN
et al
Human lymphocyte-specific pp52 gene is a member of a highly conserved dispersed family.
Genomics
1993
15
515
520
41
Ichijo
H
Nishida
E
Irie
K
et al
Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways.
Science
1997
275
90
94
42
Davis
RJ
Signal transduction by the JNK group of MAP kinases.
Cell
2000
103
239
252
43
Dong
C
Davis
RJ
Flavell
RA
MAP kinases in the immune response.
Annu Rev Immunol
2002
20
55
72
44
Sun
X
Lee
J
Navas
T
Baldwin
DT
Stewart
TA
Dixit
VM
RIP3, a novel apoptosis-inducing kinase.
J Biol Chem
1999
274
16871
16875
45
Yu
PW
Huang
BC
Shen
M
et al
Identification of RIP3, a RIP-like kinase that activates apoptosis and NFkappaB.
Curr Biol
1999
9
539
542
46
Yang
Y
Hu
W
Feng
S
Ma
J
Wu
M
RIP3 beta and RIP3 gamma, two novel splice variants of receptor-interacting protein 3 (RIP3), downregulate RIP3-induced apoptosis.
Biochem Biophys Res Commun
2005
332
181
187
47
Ma
L
Raycroft
L
Asa
D
Anderson
DC
Geng
JG
A sialoglycoprotein from human leukocytes functions as a ligand for P-selectin.
J Biol Chem
1994
269
27739
27746
48
Hidari
KI
Weyrich
AS
Zimmerman
GA
McEver
RP
Engagement of P-selectin glycoprotein ligand-1 enhances tyrosine phosphorylation and activates mitogen-activated protein kinases in human neutrophils.
J Biol Chem
1997
272
28750
28756
49
Hoffjan
S
Stemmler
S
Parwez
Q
et al
Evaluation of the toll-like receptor 6 Ser249Pro polymorphism in patients with asthma, atopic dermatitis and chronic obstructive pulmonary disease.
BMC Med Genet
2005
6
34
50
Tsan
MF
Toll-like receptors, inflammation and cancer.
Semin Cancer Biol
2006
16
32
37
51
Puthothu
B
Heinzmann
A
Is toll-like receptor 6 or toll-like receptor 10 involved in asthma genetics—or both?
Allergy
2006
61
649
650
52
Zapata
JM
Krajewska
M
Krajewski
S
et al
TNFR-associated factor family protein expression in normal tissues and lymphoid malignancies.
J Immunol
2000
165
5084
5096
53
Munzert
G
Kirchner
D
Stobbe
H
et al
Tumor necrosis factor receptor-associated factor 1 gene overexpression in B-cell chronic lymphocytic leukemia: analysis of NF-kappa B/Rel-regulated inhibitors of apoptosis.
Blood
2002
100
3749
3756
54
Siemienski
K
Peters
N
Scheurich
P
Wajant
H
Organization of the human tumour necrosis factor receptor-associated factor 1 (TRAF1) gene and mapping to chromosome 9q33–34.
Gene
1997
195
35
39
55
Tsitsikov
EN
Laouini
D
Dunn
IF
et al
TRAF1 is a negative regulator of TNF signaling: enhanced TNF signaling in TRAF1-deficient mice.
Immunity
2001
15
647
657
56
Bryce
PJ
Oyoshi
MK
Kawamoto
S
Oettgen
HC
Tsitsikov
EN
TRAF1 regulates Th2 differentiation, allergic inflammation and nuclear localization of the Th2 transcription factor, NIP45.
Int Immunol
2006
18
101
111
57
Hodzic
DM
Yeater
DB
Bengtsson
L
Otto
H
Stahl
PD
Sun2 is a novel mammalian inner nuclear membrane protein.
J Biol Chem
2004
279
25805
25812
58
Wang
Q
Du
X
Cai
Z
Greene
MI
Characterization of the structures involved in localization of the SUN proteins to the nuclear envelope and the centrosome.
DNA Cell Biol
2006
25
554
562
59
Purdue
MP
Lan
Q
Kricker
A
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
712
60
Warzocha
K
Ribeiro
P
Bienvenu
J
et al
Genetic polymorphisms in the tumor necrosis factor locus influence non-Hodgkin's lymphoma outcome.
Blood
1998
91
3574
3581
61
Juszczynski
P
Kalinka
E
Bienvenu
J
et al
Human leukocyte antigens class II and tumor necrosis factor genetic polymorphisms are independent predictors of non-Hodgkin lymphoma outcome.
Blood
2002
100
3037
3040
62
Trinchieri
G
Sher
A
Cooperation of Toll-like receptor signals in innate immune defence.
Nat Rev Immunol
2007
7
179
190
63
Nieters
A
Beckmann
L
Deeg
E
Becker
N
Gene polymorphisms in Toll-like receptors, interleukin-10, and interleukin-10 receptor alpha and lymphoma risk.
Genes Immun
2006
7
615
624
64
Liu
Y
Shepherd
EG
Nelin
LD
MAPK phosphatases: regulating the immune response.
Nat Rev Immunol
2007
7
202
212
65
Jornot
L
Petersen
H
Junod
AF
Modulation of the DNA binding activity of transcription factors CREB, NFkappaB and HSF by H2O2 and TNF alpha: differences between in vivo and in vitro effects.
FEBS Lett
1997
416
381
386
66
Gustin
JA
Pincheira
R
Mayo
LD
et al
Tumor necrosis factor activates CRE-binding protein through a p38 MAPK/MSK1 signaling pathway in endothelial cells.
Am J Physiol Cell Physiol
2004
286
C547
555
67
Witz
IP
The involvement of selectins and their ligands in tumor-progression.
Immunol Lett
2006
104
89
93
68
Blanks
JE
Moll
T
Eytner
R
Vestweber
D
Stimulation of P-selectin glycoprotein ligand-1 on mouse neutrophils activates beta 2-integrin mediated cell attachment to ICAM-1.
Eur J Immunol
1998
28
433
443
69
St-Pierre
Y
Organizing a tete-a-tete between cell adhesion molecules and extracellular proteases: a risky business that could lead to the survival of tumor cells.
Front Biosci
2005
10
1040
1049
70
Blann
AD
Gurney
D
Wadley
M
Bareford
D
Stonelake
P
Lip
GY
Increased soluble P-selectin in patients with haematological and breast cancer: a comparison with fibrinogen, plasminogen activator inhibitor and von Willebrand factor.
Blood Coagul Fibrinolysis
2001
12
43
50
71
Nash
GF
Walsh
DC
Kakkar
AK
The role of the coagulation system in tumour angiogenesis.
Lancet Oncol
2001
2
608
613
72
Lopez-Pedrera
C
Barbarroja
N
Dorado
G
Siendones
E
Velasco
F
Tissue factor as an effector of angiogenesis and tumor progression in hematological malignancies.
Leukemia
2006
20
1331
1340
73
Bennett
JS
Catella-Lawson
F
Rut
AR
et al
Effect of the Pl(A2) alloantigen on the function of beta(3)-integrins in platelets.
Blood
2001
97
3093
3099
74
Feng
D
Lindpaintner
K
Larson
MG
et al
Increased platelet aggregability associated with platelet GPIIIa PlA2 polymorphism: the Framingham Offspring Study.
Arterioscler Thromb Vasc Biol
1999
19
1142
1147
75
Szczeklik
A
Undas
A
Sanak
M
Frolow
M
Wegrzyn
W
Relationship between bleeding time, aspirin and the PlA1/A2 polymorphism of platelet glycoprotein IIIa.
Br J Haematol
2000
110
965
967
76
Vijayan
KV
Liu
Y
Dong
JF
Bray
PF
Enhanced activation of mitogen-activated protein kinase and myosin light chain kinase by the Pro33 polymorphism of integrin beta 3.
J Biol Chem
2003
278
3860
3867
77
Baron
JA
Gridley
G
Weiderpass
E
Nyren
O
Linet
M
Venous thromboembolism and cancer.
Lancet
1998
351
1077
1080
78
Garte
S
Gaspari
L
Alexandrie
AK
et al
Metabolic gene polymorphism frequencies in control populations.
Cancer Epidemiol Biomarkers Prev
2001
10
1239
1248
Sign in via your Institution