To identify genetic variants associated with outcome from chronic lymphocytic leukemia (CLL), we genotyped 977 nonsynonymous single nucleotide polymorphisms (nsSNPs) in 755 genes with relevance to cancer biology in 425 patients participating in a phase 3 trial comparing the efficacy of fludarabine, chlorambucil, and fludarabine with cyclophosphamide as first-line treatment. Selection of nsSNPs was biased toward those likely to be functionally deleterious. SNP genotypes were linked to individual patient outcome data and response to chemotherapy. The effect of genotype on progression-free survival (PFS) and overall survival (OS) was assessed by Cox regression analysis adjusting for treatment and clinico-pathologic variables. A total of 78 SNPs (51 dominantly acting and a further 27 recessively acting) were associated with PFS (9 also affecting OS) at the 5% level. These included SNPs mapping to the immune-regulation genes IL16 P434S (P = .03), IL19 S213F (P = .001), LILRA4 P27L (P = .004), KLRC4 S29I (P = .007), and CD5 V471A (P = .002); and DNA response genes POLB P242R (P = .04) and TOPBP1 S730L (P = .02), which were all independently prognostic of immunoglobulin heavy-chain variable region (IgVH) mutational status. The variants identified warrant further evaluation as promising prognostic markers of patient outcome. To facilitate the identification of prognostic markers through pooled analyses, we have made all data from our analysis publicly available.

B-cell chronic lymphocytic leukemia (B-CLL) accounts for approximately 25% of all leukemia and is the most common lymphoid malignancy in Western countries.1  Although staging systems (Binet, RAi) are useful for predicting prognosis and treatment requirements,2  there is variability in clinical outcome for patients with apparently the same stage of disease. Hence, accurate assessment of prognosis would be beneficial in the choice of specific therapeutic options, while assessment of the likelihood of response and adverse reactions to chemotherapeutic treatments would allow patient-tailored decisions on drug selection and consequent improvements in survival.

Molecular markers such as immunoglobulin heavy-chain variable region (IgVH) mutational status have been shown to greatly assist in defining patient prognosis.3-5  In addition to somatic differences, it is probable that germ-line variation also plays a role in defining individual patient outcome. Studies that have investigated this possibility have to date only evaluated a restricted number of genetic variants in a restricted number of candidate genes—polymorphisms in the coding sequence of P2×7,6 GNAS,7 SDF-1,8 MTHFR,9 p53,10  and polymorphisms in the putative promoter sequences of TNFα,11 IL-1 and IL-6,12 BCL-2,13  and BAX.14,15  However, many of these purported associations such as those in P2×7, BAX, and p53 have been unconfirmed by independent studies.16-19 

To date, most searches for polymorphic markers of prognosis have been formulated on an “educated guess”; however, without a full understanding of the biology of CLL, definition of additional “suitable” candidate genes is problematic. This makes searches on a “genome-wide” basis an attractive proposition. A further issue with many of the previously reported studies is that they have been based on small numbers of patients ascertained outside the context of a clinical trial and in addition to having limited power, are prone to bias as a consequence of survivorship and other cofounders, especially when based on retrospectively ascertained patients who have received nonuniform treatment.

We have recently conducted an association study based on an analysis of nonsynonymous single nucleotide polymorphisms (nsSNPs) in genes with a priori relevance to cancer biology to identify susceptibility alleles for CLL.20  As nsSNPs alter the encoded amino acid sequence, they have the potential to directly affect the function of expressed proteins. A total of 425 patients with CLL genotyped in this study were participants in a large phase 3 clinical trial. Linking genetic data to clinical outcome information for these patients has allowed us to test, in an unbiased fashion, the hypothesis that sequence variation defined by these nsSNPs influences the clinical behavior of CLL.

Patients

The study is based on patients entered in the UK Leukemia Research Fund (LRF) CLL-4 trial. Full details about the design and conduct of the trial are described in previously published material.21  Briefly, CLL-4 was a randomized phase 3 trial established to compare the efficacy of fludarabine, chlorambucil, and the combination of fludarabine plus cyclophosphamide as a first-line treatment for Binet stages B, C, and A-progressive CLL. The trial as a whole recruited from a total of 136 centres, primarily from the United Kingdom. Of the 777 patients entered into the trial, the current analysis is based on a random subset of 425 white patients from 110 of the participating centers which had blood samples taken for clinical diagnostic purposes and cell marker studies. Ethical approval for the study was obtained from the Multi-Center Research Ethics Committee in accordance with the Declaration of Helsinki.

Selection of candidate genes, SNP selection and genotyping

Candidate gene selection and SNP genotyping has been described previously.22  Briefly, nsSNPs mapping to candidate cancer genes were identified by interrogation of the Predicted Impact of Coding SNPs (PICS) database23  of potential functional SNPs and published work on the resequencing of DNA repair genes. To increase the likelihood of identifying susceptibility alleles, we analyzed nsSNPs in 871 genes with relevance to tumor biology, biasing selection of SNPs to those likely to directly have functional consequences. nsSNPs were genotyped across patient DNAs extracted by standard salt-lysis protocol using customized Illumina Sentrix Bead Arrays according to the manufacturer's protocols (Illumina, San Diego, CA).

To predict the impact of missense variants on protein function, we applied 2 in silico algorithms, polymorphism phenotyping (PolyPhen)24  and sifting intolerant from tolerant (SIFT).25  PolyPhen predicts the functional impact of amino acid changes by considering evolutionary conservation, physicochemical differences, and the proximity of the substitution to predicted functional domains and/or structural features. SIFT predicts the functional importance of amino acid substitutions based on the alignment of orthologous and/or paralogous protein sequences. SIFT and PolyPhen scores were classified as intolerant, potentially intolerant, borderline, or tolerant and probably damaging, possibly damaging, potentially damaging, borderline, or benign, respectively, according to the classifications proposed by Xi et al26  and Ng and Henikoff.25  Where it was not possible to derive SIFT or PolyPhen metrics, we categorized codon replacements on the basis of Grantham matrix27  according to the classification of Li et al.28 

Statistical methods

To examine for systematic biases, we assessed the deviation of the genotype frequencies in the patients from that expected under Hardy-Weinberg Equilibrium (HWE) using a quantile-quantile (Q-Q) plot and estimated an overdispersion factor λ29  by calculating the ratio of the mean of the smallest 90% of observed test statistics to the mean of the corresponding expected values.

Progression-free survival (PFS) and overall survival (OS) of patients were the end points of the analysis. The response recorded for each patient was the best achieved at any time due to first-line treatment. PFS was defined as the survival time from the date of randomization to relapse needing further therapy, progression, or death from any cause. For nonresponders and progressive disease, the date of progression was when no response or progressive disease was recorded. OS was calculated from randomization to death from any cause. Data on age at diagnosis, stage, sex, and treatment were available for all patients, and IgVH mutation status data were available for a subset of patients, enabling us to examine the significance of each SNP conditional on these covariates. Cox-regression analysis was used to estimate genotype-specific hazard ratios (HRs) and 95% confidence intervals (CIs). For each SNP genotype, HRs were generated using common allele homozygotes as the reference group. P values presented correspond to the significance of a test difference among all 3 of the genotype groups (common allele homozygote, heterozygote, and rare allele homozygote). For SNPs where 5 or fewer minor allele homozygotes were observed, minor allele homozygote genotypes were combined with heterozygotes. If this combined frequency was still 5 or less, then the SNP was removed from the analysis. HRs, 95% CIs, and associated P values under both dominant and recessive models were also generated. To assess the distribution of test statistics, we generated Q-Q plots. To investigate possible epistatic interactions, the relationship between PFS and each pair of SNPs that showed significant allelic association at the 5% level was evaluated. PFS curves were plotted using Kaplan-Meier estimates. All statistical analyses were undertaken using SAS Version 9.1 (SAS Institute, Cary, NC) and R Version 2.5.0 (The R Foundation for Statistical Computing, www.r-project.org/foundation/main.html).

Descriptive statistics

The clinicopathologic characteristics of patients studied are detailed in Table 1. Of the patients genotyped, approximately a third had been diagnosed with CLL before age 60 years, and approximately two-thirds had presented with stage B or C disease. The median follow-up time for patients was 32 months with follow-up to October, 31, 2005. There were 27% deaths in the cohort. Sex, stage at presentation, and drug therapy all significantly influenced PFS. Specifically, being female, having early-stage disease, and the combination of fludarabine and cyclophosphamide as drug therapy were all significantly associated with a more favorable prognosis (Table 2). Only age and stage at presentation significantly affected OS. For 354 (83%) of the 425 patients, we had information on IgVH mutation status. As in the parent trial, this covariate was prognostically important, being predictive of both PFS and OS (P < .001 and .002, respectively).

Table 1

Comparison of patient characteristics and details of drug treatment received by CLL-4 patients genotyped and those excluded from study

CLL-4 patients genotyped (N = 425), no. (%)CLL-4 patients excluded (N = 352), no. (%)P*
Sex    
    Male 312 (73.4) 261 (74.1) — 
    Female 113 (26.6) 91 (25.9) .8 
Age    
    Less than 60 y 127 (29.9) 127 (36.1) — 
    60 to 69 y 168 (39.5) 121 (34.4) — 
    Over 70 y 130 (30.6) 104 (29.5) .2 
Stage    
    A 104 (24.5) 87 (24.7) — 
    B 186 (43.8) 166 (47.2) — 
    C 135 (31.8) 99 (28.1) .5 
Died    
    Yes 114 (26.8) 88 (25.0) — 
    No 311 (73.2) 264 (75.0) .6 
Treatment    
    Chlorambucil 206 (48.5) 181 (51.4) — 
    Fludarabine 112 (26.4) 82 (23.3) — 
    Fludarabine and cyclophosphamide 107 (25.2) 89 (25.3) .6 
CLL-4 patients genotyped (N = 425), no. (%)CLL-4 patients excluded (N = 352), no. (%)P*
Sex    
    Male 312 (73.4) 261 (74.1) — 
    Female 113 (26.6) 91 (25.9) .8 
Age    
    Less than 60 y 127 (29.9) 127 (36.1) — 
    60 to 69 y 168 (39.5) 121 (34.4) — 
    Over 70 y 130 (30.6) 104 (29.5) .2 
Stage    
    A 104 (24.5) 87 (24.7) — 
    B 186 (43.8) 166 (47.2) — 
    C 135 (31.8) 99 (28.1) .5 
Died    
    Yes 114 (26.8) 88 (25.0) — 
    No 311 (73.2) 264 (75.0) .6 
Treatment    
    Chlorambucil 206 (48.5) 181 (51.4) — 
    Fludarabine 112 (26.4) 82 (23.3) — 
    Fludarabine and cyclophosphamide 107 (25.2) 89 (25.3) .6 

—indicates not applicable.

*

Chi-square test.

Table 2

Relationship between treatment, stage, age and sex, and PFS and OS in the patients genotyped

NOS
PFS
No. deathsO/E ratioP*Progression or deathO/E ratioP*
Treatment        
    Chlorambucil 206 48 0.87 .3 155 1.36 < .001 
    Fludarabine 112 33 1.16 — 75 1.12 — 
    Fludarabine and cyclophosphamide 107 33 1.08 — 52 0.51 — 
Stage        
    A 104 20 0.67 < .001 66 0.94 .03 
    B 186 45 0.88 — 116 0.87 — 
    C 135 49 1.5 — 100 1.28 — 
Age        
    Less than 60 y 127 21 0.51 < .001 88 1.01 .8 
    60 to 69 y 168 42 0.94 — 109 0.96 — 
    Over 70 y 130 51 1.81 — 85 1.05 — 
Sex        
    Male 312 88 1.05 .4 218 1.08 .02 
    Female 113 26 0.87 — 64 0.79 — 
NOS
PFS
No. deathsO/E ratioP*Progression or deathO/E ratioP*
Treatment        
    Chlorambucil 206 48 0.87 .3 155 1.36 < .001 
    Fludarabine 112 33 1.16 — 75 1.12 — 
    Fludarabine and cyclophosphamide 107 33 1.08 — 52 0.51 — 
Stage        
    A 104 20 0.67 < .001 66 0.94 .03 
    B 186 45 0.88 — 116 0.87 — 
    C 135 49 1.5 — 100 1.28 — 
Age        
    Less than 60 y 127 21 0.51 < .001 88 1.01 .8 
    60 to 69 y 168 42 0.94 — 109 0.96 — 
    Over 70 y 130 51 1.81 — 85 1.05 — 
Sex        
    Male 312 88 1.05 .4 218 1.08 .02 
    Female 113 26 0.87 — 64 0.79 — 

O/E ratio indicates observed versus expected ratio.

*

Log-rank test for trend.

There was no difference in the demographics, treatment, and follow-up characteristics of the 425 patients genotyped herein compared with the patients entered into CLL-4 but who were excluded from this analysis (Table 1). The observation that conventional staging and other prognosis markers were equally predictive of survival in the complete dataset and the subgroup analyzed provides further evidence that subselection of patients genotyped is unlikely to bias overall study findings.

Relationship between SNP genotype and PFS

Within the parent study, SNP call rates per sample were greater than 99% in patients. Of the 1467 SNPs submitted for analysis, 1218 (83%) were satisfactory genotyped, with mean individual SNP call rates of 99.7% Of the 1218 SNPs loci that satisfactorily genotyped, 241 were either fixed in all patient samples or were observed at subpolymorphic frequencies (ie, having minor allele frequency [MAF] of less than 1%), and hence were excluded from all analyses. Distribution of SNP genotypes was not significantly different to that expected under HWE, with λequal to 1.02 (Figure S1, available on the Blood website; see the Supplemental Materials link at the top of the online article). Of the 977 remaining SNPs mapping to the 755 genes to be analyzed, cell counts of minor allele genotypes were sufficiently infrequent to exclude 57 from the genotype and dominant analyses and 438 from the recessive model analyses. The SNPs for which genotype-survival associations were examined are detailed in Table S1 together with their associated MAFs.

As 10% to 15% of patients with CLL with progressive disease are thought to display deletions involving 17p, we examined this particular locus in detail. Genotypes for the p53 Arg72Pro (rs104252230 ) were obtained for 421 (99.1%) of the 425 patients, and the distribution of genotypes (239, 154, 28) was not significantly different to those we have previously documented in the United Kingdom population31  (1471, 1022, 197; P = .64).

After adjustment for sex, age, stage of disease at presentation, and drug regime, statistically significant associations between genotype and PFS were identified for 42 SNPs at the 5% level. A Q-Q plot of test statistics showed no large deviation from the expected distribution (Figure S2). Results of all SNPs included in the genotype-survival analysis are detailed in Table S2. On the basis of dominant and recessive models, 78 different nsSNPs were significant, 51 under a dominant model and 27 under a recessive model; none, however, were significant after adjustment for multiple testing using the conservative Bonferroni correction.

With such caveats in mind, several potentially interesting findings were observed. Of the SNPs identified to be prognostic (on the basis of nominal P ≤ .05), 2 have been established to be functional, and a further 28 SNPs are predicted to be functional (Table 3). Collectively, the SNPs predictive of PFS mapped to a restricted number of gene ontologies, and for purposes of clarity, we have restricted our commentary to the genes within pathways for which polymorphic variation in at least one SNP was associated with PFS, specifically genes involved in DNA damage-response/repair and immune regulation.

Table 3

SNPs showing significant genotypic association with PFS

GeneSymbolSNP
Reference allele (dbSNP)N*HR95% CIP
Dominantly acting alleles         
    Sec23 homolog B SEC23B H489Q — 93 0.60 0.45-0.81 .001 
    Leukocyte immunoglobulin-like receptor, subfamily A member 4 LILRA4 P27L rs2241384 285 1.44 1.12-1.85 .004 
    Chondroitin sulfate galnact-2 GALNACT-2 P479S rs2435381 237 0.70 0.55-0.89 .004 
    Cubilin (intrinsic factor-cobalamin receptor) CUBN S253F rs1801222 151 1.42 1.11-1.83 .006 
    Killer cell lectin-like receptor subfamily C, member 4 KLRC4 S29I rs1841958 195 0.72 0.57-0.91 .007 
    Phospholysine phosphohistidine inorganic pyrophosphate phosphatase LHPP R94Q rs6597801 344 1.49 1.11-1.99 .008 
    Melanoma inhibitory activity 2 MIA2 D547H rs10134365 269 1.39 1.09-1.78 .008 
    ARP10 protein ARP10 K121N rs139299 114 1.47 1.11-1.95 .008 
    Dystonin DST Q1634R rs4712138 153 1.39 1.08-1.79 .011 
    Myosin binding protein C, cardiac MYBPC3 V158M rs3729986 350 1.47 1.09-1.99 .012 
    Sema domain, immunoglobulin domain, short basic domain, secreted SEMA3C V337M rs1527482 419 3.15 1.28-7.75 .013 
    Zinc finger protein 169 ZNF169 Q583H rs12350212 344 1.46 1.08-1.97 .013 
    Serologically defined colon cancer antigen 3-like SDCCAG3L I437T rs2000746 218 0.74 0.58-0.94 .013 
    Phospholipase A2, group VII PLA2G7 A379V rs1051931 281 0.72 0.56-0.94 .014 
    Apolipoprotein E APOE R176C§ rs7412 352 1.46 1.08-1.98 .015 
    BRCA1-associated RING domain 1 BARD1 S378R rs2229571 154 0.75 0.59-0.95 .019 
    General transcription factor IIE, polypeptide 1, alpha 56 kDa GTF2E1 P366S rs3732401 387 0.56 0.34-0.91 .02 
    X-ray repair complementing defective repair in Chinese hamster cells 2 XRCC2 R188H rs3218536 354 1.44 1.06-1.95 .02 
    Excision repair cross-complementing rodent repair deficiency, complementation group ERCC6 G399D rs2228528 299 1.35 1.05-1.73 .021 
    Low-density lipoprotein receptor–related protein 4 LRP4 G1554S rs2306029 133 1.37 1.05-1.78 .021 
    Erbb2 interacting protein ERBB2IP S274L rs3213837 291 1.34 1.04-1.72 .022 
    Low-density lipoprotein–related protein 2 LRP2 L4210I rs4667591 262 1.32 1.04-1.68 .024 
    Cancer susceptibility candidate 5 CASC5 M598T rs11858113 139 0.75 0.59-0.96 .025 
    Tyrosinase TYR S192Y rs1042602 159 0.76 0.60-0.97 .027 
    Thrombospondin 1 THBS1 N700S§ rs17632786 325 0.72 0.54-0.96 .027 
    Excision repair cross-complementing rodent repair deficiency, complementation group ERCC5 H1104D rs17655 256 1.32 1.03-1.69 .028 
    Transmembrane protease, serine 4 TMPRSS4 G206V rs1941635 395 1.57 1.05-2.35 .028 
    E2F transcription factor 2 E2F2 G205R rs3218170 412 1.98 1.08-3.62 .028 
    Laminin, alpha 2 LAMA2 V1138M rs2306942 378 1.51 1.04-2.18 .028 
    Interleukin-16 (lymphocyte chemoattractant factor) IL16 P434S rs4072111 334 0.72 0.53-0.97 .031 
    Carboxypeptidase A4 CPA4 G303C rs2171492 153 0.76 0.6-0.98 .032 
    RAD18 homolog RAD18 Q302R rs373572 223 1.29 1.02-1.64 .034 
    Cytochrome P450, family 39, subfamily A, polypeptide 1 CYP39A1 P23R rs12192544 258 1.3 1.02-1.65 .034 
    Keratin 1 KRT1 A454S rs17678945 395 1.59 1.03-2.44 .035 
    Polymerase (DNA-directed), beta POLB P242R rs3136797 409 1.87 1.04-3.36 .036 
    Breast cancer 2, early onset BRCA2 T1915M rs4987117 393 1.59 1.03-2.44 .036 
    PMS2 postmeiotic segregation increased 2 PMS2 M622I rs1805324 411 0.39 0.16-0.95 .037 
    Ubiquitin specific protease 47 USP47 V75G rs11022079 205 1.28 1.01-1.63 .039 
    Zinc finger protein 311 ZNF311 K511Q rs6456880 168 1.29 1.01-1.65 .039 
    Zinc finger CCCH-type containing 3 ZC3H3 G452S rs4874147 245 1.30 1.01-1.66 .040 
    Protein tyrosine phosphatase, nonreceptor type 13 PTPN13 Y2062D rs989902 146 0.78 0.61-0.99 .041 
    Homeodomain interacting protein kinase 4 HIPK4 R302Q rs11670988 347 1.37 1.01-1.87 .042 
    RAD1 homolog (Saccharomyces pombeRAD1 E281G rs1805327 357 0.71 0.50-0.99 .042 
    Cytochrome P450, family 2, subfamily C, polypeptide 9 CYP2C9 I359L rs1057910 370 1.42 1.01-2.00 .042 
    Forkhead box N1 FOXN1 R69C rs2071587 350 1.37 1.01-1.85 .042 
    Ectonucleotide pyrophosphatase/phosphodiesterase 5 ENPP5 V171I rs6926570 117 0.76 0.58-0.99 .043 
    FRAS1-related extracellular matrix protein 2 FREM2 F1070S rs2496425 231 1.28 1.01-1.62 .044 
    Serine/threonine kinase 6 STK6 I31F rs2273535 261 0.78 0.61-0.99 .045 
    O-6-methylguanine-DNA methyltransferase MGMT L84F rs12917 327 0.75 0.56-0.99 .045 
    Carbonyl reductase 3 CBR3 V244M rs1056892 186 1.28 1.00-1.63 .046 
    Zinc finger protein 527 ZNF527 H181R rs4452075 295 0.76 0.59-1.00 .047 
Recessively acting alleles         
    Interleukin-19 IL19 S213F rs2243191 16 2.53 1.46-4.38 .001 
    Cadherin 11, type 2, OB-cadherin CDH11 T255M rs35195 47 1.73 1.22-2.45 .002 
    Cathepsin B CTSB L26V rs12338 71 0.57 0.40-0.82 .002 
    CD5 antigen (p56–62) CD5 V471A rs13328900 104 1.50 1.16-1.96 .002 
    Extra spindle poles–like 1 (S cerevisiaeESPL1 R614S rs1318648 58 1.60 1.15-2.22 .006 
    Ectonucleotide pyrophosphatase/phosphodiesterase 5 ENPP5 V171I rs6926570 86 0.64 0.46-0.88 .006 
    Glypican 1 GPC1 G500S rs2228331 32 0.48 0.28-0.81 .006 
    Matrix metalloproteinase 10 (stromelysin 2) MMP10 G65R rs17293607 2.92 1.36-6.27 .006 
    Dual specificity phosphatase 13 DUSP13 C206Y rs3088142 77 0.63 0.45-0.88 .007 
    Sucrase-isomaltase (alpha-glucosidase) SI A231T rs9283633 71 1.51 1.12-2.05 .008 
    Neural precursor cell–expressed, developmentally down-regulated 4 NEDD4 Q260R rs2303580 15 0.35 0.16-0.79 .012 
    Reticulon 3 RTN3 A6E rs11551944 2.44 1.19-5.00 .015 
    Carnitine palmitoyltransferase II CPT2 M647V rs1799822 27 1.74 1.11-2.73 .016 
    Monoacylglycerol O-acyltransferase 1 MOGAT1 S162P rs1868024 21 1.91 1.13-3.23 .016 
    Extra spindle poles–like 1 (S cerevisiaeESPL1 D25A rs6580942 44 1.58 1.09-2.29 .017 
    Topoisomerase (DNA) II binding protein 1 TOPBP1 S730L rs17301766 18 1.89 1.12-3.19 .018 
    Zinc finger protein 643 ZNF643 C13Y rs2272994 16 0.45 0.23-0.87 .019 
    SET domain, bifurcated 2 SETDB2 G117E rs7998427 64 1.45 1.05-2.00 .023 
    Zinc finger protein 573 ZNF573 A166G rs3752365 24 1.71 1.08-2.70 .024 
    A kinase (PRKA) anchor protein (yotiao) 9 AKAP9 M463I rs6964587 65 1.43 1.05-1.97 .025 
    BCL2-like 13 (apoptosis facilitator) BCL2L13 P360S rs9306198 2.68 1.09-6.60 .032 
    Dynein, axonemal, heavy polypeptide 9 DNAH9 N2195S rs3744581 23 1.68 1.03-2.72 .037 
    A kinase (PRKA) anchor protein 10 AKAP10 R249H rs2108978 63 1.41 1.02-1.94 .038 
    DEAD (Asp-Glu-Ala-Asp) box polypeptide 27 DDX27 G206V rs11908296 22 1.70 1.03-2.80 .038 
    Inhibin, beta C INHBC R322Q rs2229357 22 1.75 1.02-2.98 .041 
    Sterile alpha motif and leucine zipper containing kinase AZK ZAK S531L rs3769148 95 0.75 0.56-1.00 .047 
    Eosinophil peroxidase EPX Q122H rs11652709 35 0.64 0.41-0.99 .047 
    Four and a half LIM domains 5 FHL5 S243R rs9373985 43 0.65 0.42-1.00 .049 
GeneSymbolSNP
Reference allele (dbSNP)N*HR95% CIP
Dominantly acting alleles         
    Sec23 homolog B SEC23B H489Q — 93 0.60 0.45-0.81 .001 
    Leukocyte immunoglobulin-like receptor, subfamily A member 4 LILRA4 P27L rs2241384 285 1.44 1.12-1.85 .004 
    Chondroitin sulfate galnact-2 GALNACT-2 P479S rs2435381 237 0.70 0.55-0.89 .004 
    Cubilin (intrinsic factor-cobalamin receptor) CUBN S253F rs1801222 151 1.42 1.11-1.83 .006 
    Killer cell lectin-like receptor subfamily C, member 4 KLRC4 S29I rs1841958 195 0.72 0.57-0.91 .007 
    Phospholysine phosphohistidine inorganic pyrophosphate phosphatase LHPP R94Q rs6597801 344 1.49 1.11-1.99 .008 
    Melanoma inhibitory activity 2 MIA2 D547H rs10134365 269 1.39 1.09-1.78 .008 
    ARP10 protein ARP10 K121N rs139299 114 1.47 1.11-1.95 .008 
    Dystonin DST Q1634R rs4712138 153 1.39 1.08-1.79 .011 
    Myosin binding protein C, cardiac MYBPC3 V158M rs3729986 350 1.47 1.09-1.99 .012 
    Sema domain, immunoglobulin domain, short basic domain, secreted SEMA3C V337M rs1527482 419 3.15 1.28-7.75 .013 
    Zinc finger protein 169 ZNF169 Q583H rs12350212 344 1.46 1.08-1.97 .013 
    Serologically defined colon cancer antigen 3-like SDCCAG3L I437T rs2000746 218 0.74 0.58-0.94 .013 
    Phospholipase A2, group VII PLA2G7 A379V rs1051931 281 0.72 0.56-0.94 .014 
    Apolipoprotein E APOE R176C§ rs7412 352 1.46 1.08-1.98 .015 
    BRCA1-associated RING domain 1 BARD1 S378R rs2229571 154 0.75 0.59-0.95 .019 
    General transcription factor IIE, polypeptide 1, alpha 56 kDa GTF2E1 P366S rs3732401 387 0.56 0.34-0.91 .02 
    X-ray repair complementing defective repair in Chinese hamster cells 2 XRCC2 R188H rs3218536 354 1.44 1.06-1.95 .02 
    Excision repair cross-complementing rodent repair deficiency, complementation group ERCC6 G399D rs2228528 299 1.35 1.05-1.73 .021 
    Low-density lipoprotein receptor–related protein 4 LRP4 G1554S rs2306029 133 1.37 1.05-1.78 .021 
    Erbb2 interacting protein ERBB2IP S274L rs3213837 291 1.34 1.04-1.72 .022 
    Low-density lipoprotein–related protein 2 LRP2 L4210I rs4667591 262 1.32 1.04-1.68 .024 
    Cancer susceptibility candidate 5 CASC5 M598T rs11858113 139 0.75 0.59-0.96 .025 
    Tyrosinase TYR S192Y rs1042602 159 0.76 0.60-0.97 .027 
    Thrombospondin 1 THBS1 N700S§ rs17632786 325 0.72 0.54-0.96 .027 
    Excision repair cross-complementing rodent repair deficiency, complementation group ERCC5 H1104D rs17655 256 1.32 1.03-1.69 .028 
    Transmembrane protease, serine 4 TMPRSS4 G206V rs1941635 395 1.57 1.05-2.35 .028 
    E2F transcription factor 2 E2F2 G205R rs3218170 412 1.98 1.08-3.62 .028 
    Laminin, alpha 2 LAMA2 V1138M rs2306942 378 1.51 1.04-2.18 .028 
    Interleukin-16 (lymphocyte chemoattractant factor) IL16 P434S rs4072111 334 0.72 0.53-0.97 .031 
    Carboxypeptidase A4 CPA4 G303C rs2171492 153 0.76 0.6-0.98 .032 
    RAD18 homolog RAD18 Q302R rs373572 223 1.29 1.02-1.64 .034 
    Cytochrome P450, family 39, subfamily A, polypeptide 1 CYP39A1 P23R rs12192544 258 1.3 1.02-1.65 .034 
    Keratin 1 KRT1 A454S rs17678945 395 1.59 1.03-2.44 .035 
    Polymerase (DNA-directed), beta POLB P242R rs3136797 409 1.87 1.04-3.36 .036 
    Breast cancer 2, early onset BRCA2 T1915M rs4987117 393 1.59 1.03-2.44 .036 
    PMS2 postmeiotic segregation increased 2 PMS2 M622I rs1805324 411 0.39 0.16-0.95 .037 
    Ubiquitin specific protease 47 USP47 V75G rs11022079 205 1.28 1.01-1.63 .039 
    Zinc finger protein 311 ZNF311 K511Q rs6456880 168 1.29 1.01-1.65 .039 
    Zinc finger CCCH-type containing 3 ZC3H3 G452S rs4874147 245 1.30 1.01-1.66 .040 
    Protein tyrosine phosphatase, nonreceptor type 13 PTPN13 Y2062D rs989902 146 0.78 0.61-0.99 .041 
    Homeodomain interacting protein kinase 4 HIPK4 R302Q rs11670988 347 1.37 1.01-1.87 .042 
    RAD1 homolog (Saccharomyces pombeRAD1 E281G rs1805327 357 0.71 0.50-0.99 .042 
    Cytochrome P450, family 2, subfamily C, polypeptide 9 CYP2C9 I359L rs1057910 370 1.42 1.01-2.00 .042 
    Forkhead box N1 FOXN1 R69C rs2071587 350 1.37 1.01-1.85 .042 
    Ectonucleotide pyrophosphatase/phosphodiesterase 5 ENPP5 V171I rs6926570 117 0.76 0.58-0.99 .043 
    FRAS1-related extracellular matrix protein 2 FREM2 F1070S rs2496425 231 1.28 1.01-1.62 .044 
    Serine/threonine kinase 6 STK6 I31F rs2273535 261 0.78 0.61-0.99 .045 
    O-6-methylguanine-DNA methyltransferase MGMT L84F rs12917 327 0.75 0.56-0.99 .045 
    Carbonyl reductase 3 CBR3 V244M rs1056892 186 1.28 1.00-1.63 .046 
    Zinc finger protein 527 ZNF527 H181R rs4452075 295 0.76 0.59-1.00 .047 
Recessively acting alleles         
    Interleukin-19 IL19 S213F rs2243191 16 2.53 1.46-4.38 .001 
    Cadherin 11, type 2, OB-cadherin CDH11 T255M rs35195 47 1.73 1.22-2.45 .002 
    Cathepsin B CTSB L26V rs12338 71 0.57 0.40-0.82 .002 
    CD5 antigen (p56–62) CD5 V471A rs13328900 104 1.50 1.16-1.96 .002 
    Extra spindle poles–like 1 (S cerevisiaeESPL1 R614S rs1318648 58 1.60 1.15-2.22 .006 
    Ectonucleotide pyrophosphatase/phosphodiesterase 5 ENPP5 V171I rs6926570 86 0.64 0.46-0.88 .006 
    Glypican 1 GPC1 G500S rs2228331 32 0.48 0.28-0.81 .006 
    Matrix metalloproteinase 10 (stromelysin 2) MMP10 G65R rs17293607 2.92 1.36-6.27 .006 
    Dual specificity phosphatase 13 DUSP13 C206Y rs3088142 77 0.63 0.45-0.88 .007 
    Sucrase-isomaltase (alpha-glucosidase) SI A231T rs9283633 71 1.51 1.12-2.05 .008 
    Neural precursor cell–expressed, developmentally down-regulated 4 NEDD4 Q260R rs2303580 15 0.35 0.16-0.79 .012 
    Reticulon 3 RTN3 A6E rs11551944 2.44 1.19-5.00 .015 
    Carnitine palmitoyltransferase II CPT2 M647V rs1799822 27 1.74 1.11-2.73 .016 
    Monoacylglycerol O-acyltransferase 1 MOGAT1 S162P rs1868024 21 1.91 1.13-3.23 .016 
    Extra spindle poles–like 1 (S cerevisiaeESPL1 D25A rs6580942 44 1.58 1.09-2.29 .017 
    Topoisomerase (DNA) II binding protein 1 TOPBP1 S730L rs17301766 18 1.89 1.12-3.19 .018 
    Zinc finger protein 643 ZNF643 C13Y rs2272994 16 0.45 0.23-0.87 .019 
    SET domain, bifurcated 2 SETDB2 G117E rs7998427 64 1.45 1.05-2.00 .023 
    Zinc finger protein 573 ZNF573 A166G rs3752365 24 1.71 1.08-2.70 .024 
    A kinase (PRKA) anchor protein (yotiao) 9 AKAP9 M463I rs6964587 65 1.43 1.05-1.97 .025 
    BCL2-like 13 (apoptosis facilitator) BCL2L13 P360S rs9306198 2.68 1.09-6.60 .032 
    Dynein, axonemal, heavy polypeptide 9 DNAH9 N2195S rs3744581 23 1.68 1.03-2.72 .037 
    A kinase (PRKA) anchor protein 10 AKAP10 R249H rs2108978 63 1.41 1.02-1.94 .038 
    DEAD (Asp-Glu-Ala-Asp) box polypeptide 27 DDX27 G206V rs11908296 22 1.70 1.03-2.80 .038 
    Inhibin, beta C INHBC R322Q rs2229357 22 1.75 1.02-2.98 .041 
    Sterile alpha motif and leucine zipper containing kinase AZK ZAK S531L rs3769148 95 0.75 0.56-1.00 .047 
    Eosinophil peroxidase EPX Q122H rs11652709 35 0.64 0.41-0.99 .047 
    Four and a half LIM domains 5 FHL5 S243R rs9373985 43 0.65 0.42-1.00 .049 

HRs adjusted for age, sex, stage of disease, and treatment.

db SNP indicates database of single nucleotide polymorphisms.

*

Number of patients with at-risk genotype.

Significant after adjustment for IgVH mutation status.

Predicted to be intolerant/potentially intolerant, damaging/probably damaging, or radical/moderately radical based on SIFT/POLYPHEN/Grantham scores.

§

Established to be functional.

DNA damage-response genes

DNA polymerase beta (POLB; MIM 17476032 ) performs base excision repair required for DNA maintenance, replication, recombination, and drug resistance. Under the Cox proportional hazards model, carrier status for POLB P242R variant genotype was significantly associated with a poorer PFS (HR = 1.87; 95% CI: 1.04-3.36; P = .04). PFS (3-year) for carriers of the wild-type genotype was 22.2% (95% CI: 7.4%-66.5%) compared with 32.7% (95% CI: 27.9%-38.4%) for those with the wild-type genotype (Figure 1).

Figure 1

Kaplan-Meier curves for POLB P242R. PFS curves associated with wild-type genotype are shown as a broken line. The solid line depicts survival curve for those with at risk variant genotype.

Figure 1

Kaplan-Meier curves for POLB P242R. PFS curves associated with wild-type genotype are shown as a broken line. The solid line depicts survival curve for those with at risk variant genotype.

Close modal

Genes participating in homologous recombination include breast cancer 2 (BRCA2; MIM 600185) and x-ray repair complementing defective repair in Chinese hamster cells (XRCC2; MIM 600375), which encodes a member of the RecA/Rad51-related protein family. Carrier status for BRCA2 T1915M and XRCC2 R188H variant genotypes were associated with poorer PFS (HR = 1.59; 95% CI: 1.03-2.44; P = .04 and HR = 1.44; 95% CI: 1.06-1.95; P = .02, respectively). Patients with the BRCA2 T1915M wild-type genotype had a 3-year PFS of 33.6% (95% CI: 28.6%-39.4%), whereas the 3-year PFS was 17.8% (95% CI: 6.9%-45.9%) in carriers of wild-type genotype (Figure 2).

Figure 2

PFS curves for BRCA2 T1915M. Lines are as defined in Figure 1.

Figure 2

PFS curves for BRCA2 T1915M. Lines are as defined in Figure 1.

Close modal

Nucleotide excision repair is the major cellular system that repairs bulky DNA adducts caused by mutagens and chemotherapy. This form of DNA repair is in part performed by the proteins encoded by Chinese hamster groups 5 and 6 (ERCC5; MIM 133530 and ERCC6; MIM 609413). Carrier status for ERCC5 H1104D and ERCC6 G399D variant genotypes were both associated with a less favorable PFS (HR = 1.32, 95% CI: 1.03-1.69; P = .03 and HR = 1.35, 95% CI: 1.05-1.73; P = .02). For ERCC5 H1104D, carriers with the wild-type genotype had a 3-year PFS of 13.8% (95% CI: 5.1%-37.7%) compared with 33.9% (95% CI: 29.0%-39.7%) for noncarriers (Figure 3).

Figure 3

PFS curves for ERCC5 H1104D. Lines are as defined in Figure 1.

Figure 3

PFS curves for ERCC5 H1104D. Lines are as defined in Figure 1.

Close modal

Other DNA damage-response/repair gene SNPs displaying an association with PFS included those mapping to postmeiotic segregation increased 2 (PMS2; MIM 600259), which participates in mismatch repair; methylguanine-DNA methyltransferase (MGMT; MIM 156569); and topoisomerase (DNA) II binding protein 1 (TOP53; MIM #07760), with carrier status for PMS2 M622I and MGMT L84F being associated with a better prognosis than those with wild-type alleles (HR = 0.39, 95% CI: 0.16-0.95; P = .04 and HR = 0.75, 95% CI: 0.66-0.99; P = .04). Conversely, homozygosity for TOPBP1 S730L variant genotype was associated with a poorer outlook (HR = 1.89, 95% CI: 1.12-3.19; P = .02).

Immune regulation genes

Preeminent among immune regulation pathway SNPs were those mapping to IL-19 (IL19; MIM 605687) and IL-16 genes (IL16; MIM 603035). Although based on relatively small numbers, homozygosity for IL19 S213F variant genotype was associated with a worse PFS (HR = 2.53, 95% CI: 1.46-4.38; P = .001). Conversely, carrier status for IL16 P434S was associated with a more favorable prognosis (HR = 0.72, 95% CI: 0.53-0.97; P = .03); 3-year PFS was 47.3% (95% CI: 37.1%-60.3%) compared with 28.4% (95% CI: 23.4%-34.6%; Figure 4). Under the recessive model, homozygosity for lymphocyte antigen CD5 (MIM 153340) V471A variant genotype was associated with a poorer prognosis (HR = 1.50; 95% CI: 1.16-1.96; P = .002). Also associated with poorer PFS were carriers of leukocyte immunoglobulin-like receptor, subfamily A member 4 (LILRA4; MIM 607517) P27L variant genotype (HR = 1.44, 95% CI: 1.12-1.85; P = .004); 3-year PFS was 23.3% (95% CI: 16.2%-33.6%) compared with 36.4% (95% CI: 30.6%-43.3%; Figure 5) and sema domain, immunoglobulin domain (Ig) short basic domain, secreted, 3C (SEMA3C; MIM 602645) V337M genotype (HR = 3.15, 95% CI: 1.28-7.75; P = .01). In contrast, carrier status for killer cell lectin-like receptor subfamily C, member 4 (KLRC4; MIM 602893) S29I variant genotype was associated with a better PFS (HR = 0.72, 95% CI: 0.57-0.91; P = .007); 3-year PFS was 38.2% (95% CI: 31.4%-46.5%) compared with 26.1% (95% CI: 20.0%-34.0%; Figure 6).

Figure 4

PFS curves for IL16 P434S. Lines are as defined in Figure 1.

Figure 4

PFS curves for IL16 P434S. Lines are as defined in Figure 1.

Close modal
Figure 5

PFS curves for LILRA4 P27L. Lines are as defined in Figure 1.

Figure 5

PFS curves for LILRA4 P27L. Lines are as defined in Figure 1.

Close modal
Figure 6

PFS curves for KLRC4 S29I. Lines are as defined in Figure 1.

Figure 6

PFS curves for KLRC4 S29I. Lines are as defined in Figure 1.

Close modal

Relationship between SNP genotype and OS

A total of 9 of the 78 SNPs associated with PFS also showed an association with OS: SEC23B H489Q, GALNACT-2 P479S, ENPP5 V171I, SEMA3C V337M, GTF2E1 P366S, INHBC R322, LAMA2 V1138M, ZNF527 H181R, and BRCA2 T1915M.

Impact of SNP genotype adjusting for IgVH mutational status

For 354 (83%) of the 425 patients, we had information on IgVH mutation status, allowing us to evaluate the impact of polymorphic variation adjusting for this significant covariate. In a multivariate analysis incorporating information on IgVH mutation status, 38 of the 78 SNPs were independently prognostic (Table 3). Among these 38 SNPs were 5 mapping to immune regulation genes IL16 P434S, IL19 S213F, LILRA4 P27L, KLRC4 S29I, and CD5 V471A and 2 within DNA response genes POLB P242R and TOPBP1 S730L.

Relationship between SNP genotypes and chemotherapy

In view of the differences in pharmacology of chlorambucil, fludarabine, and cyclophosphamide, we examined for potential interactive effects between SNPs and PFS for the different drugs separately. Table S3 details the relationship between SNPs significant at the 5% level and PFS. A total of 5 SNPs associated with PFS were common to patients treated with chlorambucil or fludarabine (DST L22S, LILRA4 P27L, SEC23B H489Q, XRCC2 R188H, and ZAK S531L), 3 SNPs were common to patients treated with either chlorambucil or fludarabine with cyclophosphamide (APBB3 C236R, ENPPS I171V, and C21orf57 S2L), and 4 SNPs were common to patients treated with either fludarabine or fludarabine with cyclophosphamide (DDX27 G206V, DPYD S534N, WNT16 G72R, and DHX16 D566G).

Using SNP genotype data generated on 425 patients within the CLL-4 trial, we have sought to identify a number of promising genetic variants with prognostic potential. A major strength of our study is its large size and the fact that we have analyzed patients entered into a phase 3 randomized trial, thereby minimizing bias. While not all of the patients entered into CLL-4 trial were analyzed, there were no salient differences in the demography of patients genotyped and those excluded from the analysis; hence, it is unlikely that any spurious biases will have influenced study findings. Furthermore, our analysis is unlikely to be confounded by population stratification. We have been able to adjust for the major molecular covariate, IgVH mutation status, and identify a number of SNPs independently predictive of PFS, including variants of IL16, IL19, CD5, and POLB. We did not, however, find evidence that MTHFR A222V or p53 Arg72Pro genotype influences prognosis as previously purported.

Despite the strong biologic plausibility for several individual associations as discussed herein, and the fact that our study is large compared with contemporaneous ones, these types of analyses are prone to false discovery as a consequence of multiple comparisons. Hence, although the signal generated from our gene level analyses suggests that we observed an approximately 8% greater signal over that expected on the basis of the dominant model, our nsSNPs findings are consistent with chance after adjusting for the number of tests used.

Although our observations must be considered preliminary and hypothesis-generating, it is intriguing that a large proportion of the SNPs we have identified as having prognostic relevance resides in genes mediating the immune response, extracellular matrix interactions, or DNA damage-response/repair. Especially relevant to CLL was the observation that variation in CD5 defined by the V471I polymorphism was a determinant of PFS. Although in part speculative, through interrogation of the Pathway Architecture program (Stratagene, La Jolla, CA), 30 (38%) of the SNPs associated with prognosis were found within interrelated genes encoding pivotal components of the DNA damage-response, immune regulation, and cell-signaling pathways (Figure 7). Biologically, it is eminently plausible that variation in these genes will affect prognosis in the case of CD5 and the interleukin genes, as levels of interleukins have been documented to influence clinical behavior.34,35  Furthermore, defects in the TP53 tumor suppressor gene pathway are known to be important in CLL, as p53 inactivation is associated with aggressive disease.36,37  It is intriguing that we identified an association with APOE, as the expressed protein has cytostatic and cytotoxic activity; moreover, apolipoprotein levels and activity of aplolipoprotein receptors have been linked to prognosis in B-CLL.38-40  The prior probability of identifying a significant association with prognosis for a series of SNPs mapping to a single DNA repair pathway is intuitively small, thereby providing a measure of robustness to our observations. However, ultimately, replication in independent studies is required to either validate or refute study findings.

Figure 7

Interrelationships between genes involved in the DNA damage-response, immune regulation, and cell-signaling pathways for SNPs associated with PFS in CLL. Interactions were established using Pathway Assist software and are color-coded: blue indicates expression; gray, regulation; and red, protein binding. Supporting publications are indicated with the corresponding National Center for Biotechnology Information (NCBI) Entrez PubMed ID in parentheses. 1, binding (14988562); 2, binding (11375975); 3, binding (15525513); 4, binding (11799067); 5, binding (12808131); 6, regulation (14744757); 7, binding (11279041); 8, binding (15735755); 9, regulation (10490955); 10, promoter binding (10942771); 11, promoter binding (1480019); 12, promoter binding (8018731); 13, binding (7498414); 14, regulation (10487844); and 15, regulation (11595644).

Figure 7

Interrelationships between genes involved in the DNA damage-response, immune regulation, and cell-signaling pathways for SNPs associated with PFS in CLL. Interactions were established using Pathway Assist software and are color-coded: blue indicates expression; gray, regulation; and red, protein binding. Supporting publications are indicated with the corresponding National Center for Biotechnology Information (NCBI) Entrez PubMed ID in parentheses. 1, binding (14988562); 2, binding (11375975); 3, binding (15525513); 4, binding (11799067); 5, binding (12808131); 6, regulation (14744757); 7, binding (11279041); 8, binding (15735755); 9, regulation (10490955); 10, promoter binding (10942771); 11, promoter binding (1480019); 12, promoter binding (8018731); 13, binding (7498414); 14, regulation (10487844); and 15, regulation (11595644).

Close modal

Our observation that polymorphic variation in a number of the DNA repair genes (ERCC5, BRCA2, and POLB) influences survival of patients with cancer is not without precedent. Outside the context of CLL variation, ERCC5 D1104H has been reported to influence esophageal41  and lung42  cancer, and POLB P242R has been reported to influence lung cancer prognosis42  in the same manner, suggesting generic effects on outcome from malignancy.

While we have evaluated only nsSNPs with a higher probability of being directly causal, there is currently direct evidence for functionality only for a minority, such as APOE R176C and THBS1 N700S.43,44  Evidence for others is provided by in silico predictions regarding the consequences of amino acid changes. While such predictions have been shown to successfully categorize a high proportion of amino acid substitutions in benchmarking studies,26,45,46  it is acknowledged that assigning functionality to SNPs on the basis of metrics is in essence speculative. Moreover, it is probable that some associations are a consequence of linkage disequilibrium with causal variants.

As the patients analyzed in our study were part of a large clinical trial, this has provided a unique opportunity to examine differential effects of SNP on PFS according to specific chemotherapy. On the basis that chlorambucil and cyclophosphamide are both alkylating agents, we have looked for SNPs common to both patient groups. Although variants such as XRCC2 R188H are attractive candidates for conferring differential response to treatment, there are currently no data for the role for variation in this or the other genes identified as a determinant of drug response. Interestingly, the S534N variant of dihydropyrimidine dehydrogenase (DPYD), shown to influence response to fludarabine, was related to PFS (P = .03 and .002 for PFS in fludarabine-treated patients and fludarabine plus cyclophosphamide-treated patients, respectively). DPYD is the initial and rate-limiting factor in the pathway of uracil and thymidine catabolism, with genetic deficiency being well recognized to result in an error in pyrimidine metabolism.47  In addition, cyclophosphamide has been shown to augment the tumor effect of pyrimidine agents such as 5-fluorouracil through inhibition of DPYD.48  In this respect, although fludarabine acts as a pyrimidine analog, it is interesting that a comparable effect was observed.

Here, we have restricted our search for SNPs associated with disease outcome by assessing those which have the potential to affect the behavior of the expressed protein through change in the amino sequence. Epigenetic silencing of DAPK1 by promoter methylation has recently been shown to occur in most patients with CLL49 ; hence, expression of this gene is a key player in CLL. Intriguingly, a rare variant in the promoter of DAPK1 was demonstrated to affect DAPK1 expression.49  It is therefore highly probable that in addition to nsSNPs, noncoding variants influencing expression of key genes such as DAPK1 will also be determinants of patient outcome and worthy of evaluation.

Our study provides evidence that inherited variation influences the clinical outcome from CLL and potentially provides additional insight into the biological determinants of prognosis. Furthermore, it also serves to highlight the statistical problem of searching for genetic associations when the impact of any variant is likely to be at best modest. Hence, stipulating significance levels of less than 10−5 for an analysis of clinical trial data is unrealistic, because to have 80% power to demonstrate a 5% difference in survival, which is clinically relevant, requires at least 4800 patient samples to be analyzed, even if the frequency of the at risk genotype is 50%. Moreover, imposing very stringent statistical thresholds can create the serious issue of type II errors. To facilitate the identification of prognostic markers through pooled analyses, we have therefore made data from our study publicly accessible (Tables S1Table S2. Results of all SNPs included in the genotype-survival analysis (XLS, 511 KB)S3, Figures S1,S2).

Finally, although germ-line variants are unlikely to replace staging schemes and conventional markers such as IgVH mutation status in the short term, they have potential to assist in distinguishing different outcome patterns among patients with the same stage of disease where 10% differences are clinically relevant, thereby opening up the possibility of a rational, targeted approach to treatment based on a combination of genotype and tumor characteristics of a patient.

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 are grateful to all patients and clinicians participating in the LRF CLL4 trial. We thank Emily Webb for additional statistical analyses and Peter Broderick for his expertise in use of the Pathway Assist Software.

This work was undertaken primarily with grant support from Leukemia Research. Additional funding was provided by the Arabib Foundation and Cancer Research UK.

Contribution: G.S.S. designed research, performed genetic analyses, and analyzed and interpreted data; R.W. and S.R. performed statistical analyses; D.G.O. performed hematologic assays; D.C. designed and conducted the clinical trial; and R.H. designed research, performed research, analyzed and interpreted data, and drafted the manuscript.

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

Correspondence: Richard S. Houlston, Section of Cancer Genetics, Institute of Cancer Research, 15 Cotswald Road, Sutton, Surrey SM2 5NG, UK; e-mail: richard.houlston@icr.ac.uk.

1
Stevenson
 
FK
Caligaris-Cappio
 
F
Chronic lymphocytic leukemia: revelations from the B-cell receptor.
Blood
2004
, vol. 
103
 (pg. 
4389
-
4395
)
2
Binet
 
JL
Prognostic factors in chronic lymphocytic leukaemia.
Haematologica
1999
, vol. 
84
 (pg. 
96
-
97
)
3
Dohner
 
H
Stilgenbauer
 
S
Benner
 
A
, et al. 
Genomic aberrations and survival in chronic lymphocytic leukemia.
N Engl J Med
2000
, vol. 
343
 (pg. 
1910
-
1916
)
4
Hamblin
 
TJ
Davis
 
Z
Gardiner
 
A
Oscier
 
DG
Stevenson
 
FK
Unmutated Ig V(H) genes are associated with a more aggressive form of chronic lymphocytic leukemia.
Blood
1999
, vol. 
94
 (pg. 
1848
-
1854
)
5
Del Poeta
 
G
Maurillo
 
L
Venditti
 
A
, et al. 
Clinical significance of CD38 expression in chronic lymphocytic leukemia.
Blood
2001
, vol. 
98
 (pg. 
2633
-
2639
)
6
Thunberg
 
U
Tobin
 
G
Johnson
 
A
, et al. 
Polymorphism in the P2X7 receptor gene and survival in chronic lymphocytic leukaemia.
Lancet
2002
, vol. 
360
 (pg. 
1935
-
1939
)
7
Frey
 
UH
Nuckel
 
H
Sellmann
 
L
, et al. 
The GNAS1 T393C polymorphism is associated with disease progression and survival in chronic lymphocytic leukemia.
Clin Cancer Res
2006
, vol. 
12
 (pg. 
5686
-
5692
)
8
Pemberton
 
NC
Paneesha
 
S
Hiller
 
L
, et al. 
The SDF-1 G > A polymorphism at position 801 plays no role in multiple myeloma but may contribute to an inferior cause-specific survival in chronic lymphocytic leukemia.
Leuk Lymphoma
2006
, vol. 
47
 (pg. 
1239
-
1244
)
9
Nuckel
 
H
Frey
 
UH
Durig
 
J
Duhrsen
 
U
Siffert
 
W
Methylenetetrahydrofolate reductase (MTHFR) gene 677C>T and 1298A>C polymorphisms are associated with differential apoptosis of leukemic B cells in vitro and disease progression in chronic lymphocytic leukemia.
Leukemia
2004
, vol. 
18
 (pg. 
1816
-
1823
)
10
Kochethu
 
G
Delgado
 
J
Pepper
 
C
, et al. 
Two germ line polymorphisms of the tumour suppressor gene p53 may influence the biology of chronic lymphocytic leukaemia.
Leuk Res
2006
, vol. 
30
 (pg. 
1113
-
1118
)
11
Au
 
WY
Fung
 
A
Wong
 
KF
Chan
 
CH
Liang
 
R
Tumor necrosis factor alpha promoter polymorphism and the risk of chronic lymphocytic leukemia and myeloma in the Chinese population.
Leuk Lymphoma
2006
, vol. 
47
 (pg. 
2189
-
2193
)
12
Hulkkonen
 
J
Vilpo
 
J
Vilpo
 
L
Koski
 
T
Hurme
 
M
Interleukin-1 beta, interleukin-1 receptor antagonist and interleukin-6 plasma levels and cytokine gene polymorphisms in chronic lymphocytic leukemia: correlation with prognostic parameters.
Haematologica
2000
, vol. 
85
 (pg. 
600
-
606
)
13
Nuckel
 
H
Frey
 
UH
Bau
 
M
, et al. 
Association of a novel regulatory polymorphism (−938C>A) in the BCL2 gene promoter with disease progression and survival in chronic lymphocytic leukemia.
Blood
2007
, vol. 
109
 (pg. 
290
-
297
)
14
Saxena
 
A
Moshynska
 
O
Sankaran
 
K
Viswanathan
 
S
Sheridan
 
DP
Association of a novel single nucleotide polymorphism, G(−248)A, in the 5′-UTR of BAX gene in chronic lymphocytic leukemia with disease progression and treatment resistance.
Cancer Lett
2002
, vol. 
187
 (pg. 
199
-
205
)
15
Starczynski
 
J
Pepper
 
C
Pratt
 
G
, et al. 
Common polymorphism G(−248)A in the promoter region of the bax gene results in significantly shorter survival in patients with chronic lymphocytic Leukemia once treatment is initiated.
J Clin Oncol
2005
, vol. 
23
 (pg. 
1514
-
1521
)
16
Starczynski
 
J
Pepper
 
C
Pratt
 
G
, et al. 
The P2X7 receptor gene polymorphism 1513 A->C has no effect on clinical prognostic markers, in vitro sensitivity to fludarabine, Bcl-2 family protein expression or survival in B-cell chronic lymphocytic leukaemia.
Br J Haematol
2003
, vol. 
123
 (pg. 
66
-
71
)
17
Nuckel
 
H
Frey
 
UH
Durig
 
J
Duhrsen
 
U
Siffert
 
W
1513A/C polymorphism in the P2X7 receptor gene in chronic lymphocytic leukemia: absence of correlation with clinical outcome.
Eur J Haematol
2004
, vol. 
72
 (pg. 
259
-
263
)
18
Sturm
 
I
Bosanquet
 
AG
Hummel
 
M
Dorken
 
B
Daniel
 
PT
In B-CLL, the codon 72 polymorphic variants of p53 are not related to drug resistance and disease prognosis.
BMC Cancer
2005
, vol. 
5
 pg. 
105
 
19
Skogsberg
 
S
Tobin
 
G
Krober
 
A
, et al. 
The G(−248)A polymorphism in the promoter region of the Bax gene does not correlate with prognostic markers or overall survival in chronic lymphocytic leukemia.
Leukemia
2006
, vol. 
20
 (pg. 
77
-
81
)
20
Rudd
 
MF
Sellick
 
GS
Webb
 
EL
Catovsky
 
D
Houlston
 
RS
Variants in the ATM-BRCA2-CHEK2 axis predispose to chronic lymphocytic leukemia.
Blood
2006
, vol. 
108
 (pg. 
638
-
644
)
21
Catovsky
 
D
Richards
 
S
Matutes
 
E
, et al. 
Assessment of fludarabine plus cyclophosphamide for patients with chronic lymphocytic leukaemia (the LRF CLL4 Trial): a randomised controlled trial.
Lancet
2007
, vol. 
370
 (pg. 
230
-
239
)
22
Rudd
 
MF
Williams
 
RD
Webb
 
EL
Schmidt
 
S
Sellick
 
GS
Houlston
 
RS
The predicted impact of coding single nucleotide polymorphisms database.
Cancer Epidemiol Biomarkers Prev
2005
, vol. 
14
 (pg. 
2598
-
2604
)
24
Ramensky
 
V
Bork
 
P
Sunyaev
 
S
Human non-synonymous SNPs: server and survey.
Nucleic Acids Res
2002
, vol. 
30
 (pg. 
3894
-
3900
)
25
Ng
 
PC
Henikoff
 
S
Predicting deleterious amino acid substitutions.
Genome Res
2001
, vol. 
11
 (pg. 
863
-
874
)
26
Xi
 
T
Jones
 
IM
Mohrenweiser
 
HW
Many amino acid substitution variants identified in DNA repair genes during human population screenings are predicted to impact protein function.
Genomics
2004
, vol. 
83
 (pg. 
970
-
979
)
27
Grantham
 
R
Amino acid difference formula to help explain protein evolution.
Science
1974
, vol. 
185
 (pg. 
862
-
864
)
28
Li
 
WH
Wu
 
CI
Luo
 
CC
Nonrandomness of point mutation as reflected in nucleotide substitutions in pseudogenes and its evolutionary implications.
J Mol Evol
1984
, vol. 
21
 (pg. 
58
-
71
)
29
Clayton
 
DG
Walker
 
NM
Smyth
 
DJ
, et al. 
Population structure, differential bias and genomic control in a large-scale, case-control association study.
Nat Genet
2005
, vol. 
37
 (pg. 
1243
-
1246
)
30
National Center for Biotechnology Information
Entrez SNP.
Accessed July 2007 
31
Webb
 
EL
Rudd
 
MF
Sellick
 
GS
, et al. 
Search for low penetrance alleles for colorectal cancer through a scan of 1467 non-synonymous SNPs in 2575 cases and 2707 controls with validation by kin-cohort analysis of 14 704 first-degree relatives.
Hum Mol Genet
2006
, vol. 
15
 (pg. 
3263
-
3271
)
32
National Center for Biotechnology Information
Online Mendelian Inheritance in Man (OMIM).
Accessed July 2007 
33
National Center for Biotechnology Information
Entrez PubMed.
Accessed July 2007 
34
Wierda
 
WG
Johnson
 
MM
Do
 
KA
, et al. 
Plasma interleukin 8 level predicts for survival in chronic lymphocytic leukaemia.
Br J Haematol
2003
, vol. 
120
 (pg. 
452
-
456
)
35
Fayad
 
L
Keating
 
MJ
Reuben
 
JM
, et al. 
Interleukin-6 and interleukin-10 levels in chronic lymphocytic leukemia: correlation with phenotypic characteristics and outcome.
Blood
2001
, vol. 
97
 (pg. 
256
-
263
)
36
Stankovic
 
T
Hubank
 
M
Cronin
 
D
, et al. 
Microarray analysis reveals that TP53- and ATM-mutant B-CLLs share a defect in activating proapoptotic responses after DNA damage but are distinguished by major differences in activating prosurvival responses.
Blood
2004
, vol. 
103
 (pg. 
291
-
300
)
37
Guarini
 
A
Gaidano
 
G
Mauro
 
FR
, et al. 
Chronic lymphocytic leukemia patients with highly stable and indolent disease show distinctive phenotypic and genotypic features.
Blood
2003
, vol. 
102
 (pg. 
1035
-
1041
)
38
Juliusson
 
G
Vitols
 
S
Impaired low-density lipoprotein receptor activity in chronic B-lymphocytic leukaemia cells.
Eur J Haematol
1988
, vol. 
40
 (pg. 
18
-
24
)
39
Chapman-Shimshoni
 
D
Yuklea
 
M
Radnay
 
J
Shapiro
 
H
Lishner
 
M
Simvastatin induces apoptosis of B-CLL cells by activation of mitochondrial caspase 9.
Exp Hematol
2003
, vol. 
31
 (pg. 
779
-
783
)
40
Bomstein
 
Y
Yuklea
 
M
Radnay
 
J
, et al. 
The antiapoptotic effects of blood constituents in patients with chronic lymphocytic leukemia.
Eur J Haematol
2003
, vol. 
70
 (pg. 
290
-
295
)
41
Wu
 
X
Gu
 
J
Wu
 
TT
, et al. 
Genetic variations in radiation and chemotherapy drug action pathways predict clinical outcomes in esophageal cancer.
J Clin Oncol
2006
, vol. 
24
 (pg. 
3789
-
3798
)
42
Matakidou
 
A
el Galta
 
R
Webb
 
E
, et al. 
Genetic variation in the DNA repair genes is predictive of outcome in lung cancer
Hum Mol Genet
2007
, vol. 
6
 (pg. 
2333
-
2340
)
43
Stenina
 
OI
Ustinov
 
V
Krukovets
 
I
Marinic
 
T
Topol
 
EJ
Plow
 
EF
Polymorphisms A387P in thrombospondin-4 and N700S in thrombospondin-1 perturb calcium binding sites.
FASEB J
2005
, vol. 
19
 (pg. 
1893
-
1895
)
44
Davignon
 
J
Gregg
 
RE
Sing
 
CF
Apolipoprotein E polymorphism and atherosclerosis.
Arteriosclerosis
1988
, vol. 
8
 (pg. 
1
-
21
)
45
Savas
 
S
Kim
 
DY
Ahmad
 
MF
Shariff
 
M
Ozcelik
 
H
Identifying functional genetic variants in DNA repair pathway using protein conservation analysis.
Cancer Epidemiol Biomarkers Prev
2004
, vol. 
13
 (pg. 
801
-
807
)
46
Sunyaev
 
S
Ramensky
 
V
Bork
 
P
Towards a structural basis of human non-synonymous single nucleotide polymorphisms.
Trends Genet
2000
, vol. 
16
 (pg. 
198
-
200
)
47
Johnson
 
MR
Wang
 
K
Diasio
 
RB
Profound dihydropyrimidine dehydrogenase deficiency resulting from a novel compound heterozygote genotype.
Clin Cancer Res
2002
, vol. 
8
 (pg. 
768
-
774
)
48
Nio
 
Y
Iguchi
 
C
Kodama
 
H
, et al. 
Cyclophosphamide augments the anti-tumor efficacy of uracil and tegafur by inhibiting dihydropyrimidine dehydrogenase.
Oncol Rep
2007
, vol. 
17
 (pg. 
153
-
159
)
49
Raval
 
A
Tanner
 
SM
Byrd
 
JC
, et al. 
Downregulation of death-associated protein kinase 1 (DAPK1) in chronic lymphocytic leukemia.
Cell
2007
, vol. 
129
 (pg. 
879
-
890
)
Sign in via your Institution