ABO blood groups and pancreatic cancer risk and survival: Results from the PANcreatic Disease ReseArch (PANDoRA) consortium

  • Authors:
    • Cosmeri Rizzato
    • Daniele Campa
    • Raffaele Pezzilli
    • Pavel Soucek
    • William Greenhalf
    • Gabriele Capurso
    • Renata Talar-Wojnarowska
    • Anette Heller
    • Krzysztof Jamroziak
    • Kay-Tee Khaw
    • Tim J. Key
    • Franco Bambi
    • Stefano Landi
    • Beatrice Mohelnikova-Duchonova
    • Ludmila Vodickova
    • Markus W. Büchler
    • Peter Bugert
    • Pavel Vodicka
    • John P. Neoptolemos
    • Jens Werner
    • Jörg D. Hoheisel
    • Andrea S. Bauer
    • Nathalia Giese
    • Federico Canzian
  • View Affiliations

  • Published online on: February 12, 2013     https://doi.org/10.3892/or.2013.2285
  • Pages: 1637-1644
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

There is strong epidemiologic evidence indicating that common genetic variability could be implicated in pancreatic cancer risk and, to date, various loci have been proposed. In particular, there is increasing evidence of the involvement of ABO gene variability and pancreatic cancer risk. In a large multicentric study of 1,028 pancreatic ductal adenocarcinoma cases and 2,257 controls in the context of the PANcreatic Disease ReseArch (PANDoRA) consortium, we investigated the suggested association with increased risk for carriers of single nucleotide polymorphisms (SNPs) determining the A or B allele in comparison with the O allele, which encodes for a non-functional enzyme. Since glycosyltransferase activity, encoded by ABO, is higher for the A1 variant compared with the A2 variant, we investigated the hypothesis that A1 carriers were at an increased risk of pancreatic cancer. In our analysis, carriers of the A1 were indeed at greater risk of developing the disease. In addition, we investigated the possible influence that genetic variability at the ABO locus may have in pancreatic cancer survival, but we observed no effect in our population.

Introduction

Pancreatic cancer is the fourth leading cause of cancer-related mortality in the European Union and in the USA, with a 5-year survival of less than 5% (1). The latter applies particularly for pancreatic ductal adenocarcinoma (PDAC), the most common form of the disease. There is no effective screening test for pancreatic cancer and metastatic disease is commonly present at initial diagnosis. Established risk factors include cigarette smoking, obesity, a medical history of diabetes type II and family history of pancreatic cancer (2,3). Moreover, it has been shown that a small proportion of pancreatic tumors arises as a result of high penetrance germline mutations in genes such as BRCA1, BRCA2, p16/CDKN2A, STK11/LKB, APC, PRSS1, SPINK, KRAS and PALB2(25). However, the very low frequency of those mutations cannot explain the bulk of genetic susceptibility to pancreatic cancer.

There is strong epidemiologic evidence indicating that common genetic variability could be implicated in the disease risk and various risk loci have been proposed. Three genome-wide association studies have been performed and resulted in several loci convincingly being associated with risk of pancreatic cancer (69). In particular, there is increasing evidence of the involvement of the ABO gene variability and pancreatic cancer risk (6,1012).

The ABO gene encodes enzymes known as glycosyltransferases which transfer specific sugar residues to a precursor substance (the H antigen) to produce the A and B antigens. Glycosylation is one of the most prevalent modifications mediated by complex enzymatic machinery, whereby glycans (sugars) are covalently attached to specific amino acid sites of proteins. Glycans have key biological functions in protein maturation and turnover, cell adhesion and trafficking and receptor binding and activation (13).

There are three major alleles at the ABO locus on chromosome 9q34: alleles A, B and O, defined by single base deletions and substitutions (single nucleotide polymorphisms; SNPs) occurring in exons 6 and 7. The A allele encodes α1→3 N-acetylgalactosaminyltransferase, which adds N-acetylgalactosamine (GalNAc) to the H antigen to form the A antigen. The B allele encodes α1→3 galactosyltransferase, which transfers galactose (Gal) to the H antigen to construct the B antigen (14). The O allele does not produce an active enzyme (14). Four SNPs at nucleotides (nt) 526, 703, 796 and 803 resulting in amino acid substitutions (Arg176Gly, Gly235Ser, Leu266Met and Gly268Ala) explain all the differences in the activity and the nucleotide-sugar donor specificity of the A and B transferases. In addition, a base substitution (rs1053878) at nt 467, resulting in an amino acid substitution (proline to leucine), distinguishes the A1 from A2 subtypes. A2 is present in approximately 20% of subjects with A blood group among Caucasians and shows an intermediate phenotype, between the ‘full’ enzymatic activity defined by the A1 allele and the nonfunctioning enzyme defined by the O allele (11,12). The carriers of the A or B allele show an increased risk of pancreatic cancer (11,12). A small number of studies have also tested the genetic variability of the locus with the aggressiveness of the disease and survival of patients, with inconsistent results (1517).

In this large multicentric study of 1,028 pancreatic cancer cases and 2,257 controls, we replicated the previously reported associations and we also investigated the possible influence of the genetic variability at the ABO locus on survival of patients with pancreatic cancer, considering also the polymorphism discriminating the A1 and A2 variants. Moreover, we performed stratified analysis considering the tumor stage in order to verify whether genetic variability could be involved in the disease prognosis.

Materials and methods

Study population

The study population was described in detail elsewhere (18,19). Briefly, 1,028 PDAC cases and 2,257 controls were collected retrospectively in five European countries in the context of the PANcreatic Disease ReseArch (PANDoRA) consortium (18). All cases were collected between 1996 and 2011. The cases included in the consortium population are defined by a confirmed diagnosis of PDAC. For each patient, information about gender and age at diagnosis was collected. Detailed information concerning overall survival (OS) was registered as well.

Comprehensive information on the control population was previously provided (18,19). Briefly, a total of 2,121 healthy controls of German, Italian, Czech and Polish origin were respectively recruited at the Institute of Transfusion Medicine, Mannheim, Germany; at the Azienda Ospedaliero-Universitaria Meyer (A.O.U. Meyer) Ospedale Pediatrico, Florence, Italy; at the Department of Molecular Biology of Cancer, Institute of Experimental Medicine, Academy of Science of the Czech Republic and First Faculty of Medicine, Charles University in Prague, Czech Republic; and at the Department of Hematology, Medical University, Lodz, Poland. British controls (N=136) were selected from healthy volunteers recruited from the general population in two cohorts in the European Prospective Investigation on Cancer (EPIC), an ongoing prospective cohort being carried out in ten European countries. The EPIC-Norfolk cohort (http://www.srl.cam.ac.uk/epic/) comprises >30,000 individuals, aged 45–75 years at recruitment, residents in Norfolk, East Anglia, UK, and recruited from general practice registers between 1993 and 1997 (20). The EPIC-Oxford cohort comprises 65,429 people aged ≥20 years and living in the UK, recruited between 1993 and 1999 (21). Characteristics of patients and controls are provided in Table I.

Table I

Characteristics of the study population.

Table I

Characteristics of the study population.

CzechGermanItalianBritishPolish





CasesControlsCasesControlsCasesControlsCasesControlsCasesControls
No.2263045021,50513918410813654151
Type of controlsBlood donorsBlood donorsBlood donorsGeneral populationBlood donors
Gender
 Female82133218697537840553582
 Male1441712828088610667811969
 Missing0020001000
Age at diagnosis/recruitment
 Mean (standard deviation)62 (10.4)59 (12.2)64 (9.8)56 (10.1)70 (9.9)49 (10.1)64 (10.4)60 (9.3)59 (9.9)67 (10.6)
 Median (25–75%)63 (56–69)59 (52–68)65 (59–71)58 (53–63)71 (63–77)42 (41–57)64 (58–72)61 (53–68)61 (53–67)68 (62–73)
Survival days
 Mean (standard deviation)298 (261)509 (431)329 (230)427 (508)330 (205)
 Median (25–75%)257 (96–402)369 (198–684)312 (120–450)298 (160–491)380 (162–520)
 Deaths15238311310620
Selection of genes and polymorphisms

We examined six SNPs on the ABO gene: rs505922, which marks the O allele (11,12), rs8176747, rs8176720, rs8176741, rs8176746, which discriminate between allele A and B, and rs1053878, which distinguishes between the A1 and A2 alleles. These SNPs account for all the functionally relevant variability at the ABO locus and predict the ABO blood groups, as shown in Table II.

Table II

ABO gene SNP selection.

Table II

ABO gene SNP selection.

Blood group

cDNAaaabSNPTagcOA1A2B

aaaaaa
26187rs8176719rs505922del (frame shift)
29399rs8176720glyglygly
467156rs1053878proleupro
526176rs7853989rs8176746argarggly
657219rs8176741hishishis
703235rs8176743rs8176746glyglyser
796266rs8176746leuleumet
803268rs8176747glyglyala
930268rs8176749rs8176746leuleuleu

a Position (nucleotide number) within the ABO cDNA.

b Position (amino acid/codon number) within the ABO protein.

c SNP that can be used as surrogate due to high linkage disequilibrium (r2>0.95 in HapMap CEU subjects).

DNA extraction and genotyping

DNA was extracted from whole blood or from frozen or paraffin-embedded pancreatic tissues of patients and controls using the Qiagen-mini kit (Qiagen, Hilden, Germany) or the AllPrep Isolation kit (Qiagen) according to the manufacturer's protocol. Genotyping was performed using an allele-specific PCR-based KASPar SNP genotyping system (KBiosciences, Hoddesdon, UK) as recommended by the manufacturer. The order of DNAs from cases and controls was randomized on PCR plates in order to ensure that an equal number of cases could be analyzed simultaneously. Detection was performed using an ABI PRISM 7900 HT sequence detection system with SDS 2.2 software (Applied Biosystems, Foster City, CA, USA). Genotyping for British controls was performed in the context of a genome-wide association study using the Human 660W-Quad BeadChip array according to manufacturer's instructions (Illumina, San Diego, CA, USA).

Statistical analysis

Hardy-Weinberg equilibrium was tested in the controls by the χ2 test. We used logistic regression for multivariate analyses to assess the main effects of the genetic polymorphisms on pancreatic cancer risk using a co-dominant inheritance model. The most common allele in the controls was assigned as the reference category. All analyses were adjusted for age, gender and country.

For survival analysis, the median follow-up time was computed with censored observations only, whereas the median survival time was calculated using data from all patients. OS was defined as the time interval between diagnosis and death (uncensored observation) or the last date when the patient was still alive (censored observation, mean follow-up time 543 days). OS was evaluated using methods for censored survival time. In particular, risk of mortality was estimated by hazard ratios (HR) and 95% confidence intervals (CI) in Cox proportional hazard models. All the analyses were performed with STATA software (StataCorp., College Station, TX, USA).

Results

In this study, we investigated two endpoints: i) to replicate the associations between 6 ABO SNPs, predictive of the ABO blood groups, and the risk of developing pancreatic cancer; and ii) to evaluate the possible associations between the same SNPs and patient survival.

We carried out genotyping in 1,029 cases and 2,280 controls. Samples of 1 case and 23 controls were excluded due to low call rates (<5 SNPs successfully genotyped), therefore we performed statistical analyses on 1,028 cases and 2,257 controls. The average call rate of the SNPs was 96.78% (range 94.48–99.13%). For 27 cases, both normal and tumor tissues were available and used for genotyping. No differences were observed (398 informative genotype comparisons). Approximately 15% of the samples were analyzed in duplicate and the concordance rate of the genotypes was >99%. In controls, the genotype distributions at all loci were within the Hardy-Weinberg equilibrium, with non-significant χ2 values (data not shown).

By examining all the SNPs separately in relation to pancreatic cancer risk, we found that rs8176741, rs8176746 and rs8176747 were associated with decreased risk of pancreatic cancer, while rs505922 was associated with increased risk of developing the disease. The association of this SNP with pancreatic cancer risk was previously evaluated in a subgroup of the patients of this study and has already been reported (19). The remaining two SNPs (rs1053878 and rs8176720) were not associated with pancreatic cancer risk. The strongest association observed was that with the T allele of the rs8176741 SNP (ORhet=0.73; 95% CI 0.58–0.90; ORcarriers=0.75; 95% CI: 0.60–0.92). The corresponding frequencies and distribution of the genotypes and the odds ratios for the association of each polymorphism with PDAC risk are summarized in Table III. We estimated the risk of pancreatic cancer according to genotype-derived ABO blood type among all the study participants, as shown in Table IV. For 1,035 German controls, information on ABO status, defined serologically (O, A, AB or B), was collected at recruitment. In 99.3% of subjects (1,028/1,035), the serological blood type and the genotype-derived blood type were identical; this proportion strongly supports the accuracy of the genotype-defined blood group alleles.

Table III

Associations between individual ABO SNPs and risk of PDAC.

Table III

Associations between individual ABO SNPs and risk of PDAC.

SNPGenotypeCasesaControlsaORb(95% CI)bP-valuePtrend
rs8176720AA4639740.168
AG44510030.97(0.82–1.14)0.693
GG1112750.84(0.65–1.1)0.205
AG+GG0.94(0.8–1.1)0.443
rs8176741CC84516890.009
CT1484000.73(0.58–0.9)0.004
TT12271.07(0.52–2.19)0.861
CT+TT0.75(0.6–0.92)0.006
rs8176746CC85618200.068
CA1584120.80(0.65–0.99)0.04
AA11251.03(0.49–2.15)0.948
CA+AA0.81(0.66–1)0.049
rs8176747GG85716970.027
CG1604050.77(0.63–0.95)0.017
CC11241.03(0.48–2.18)0.946
CG+GG0.79(0.64–0.97)0.022
rs505922TT3428600.026
CT51810551.18(0.99–1.4)0.066
CC1633361.19(0.94–1.51)0.147
CT+CC1.18(1–1.39)0.048
rs1053878AA0.538
AG86619571.05(0.84–1.33)0.659
GG1402850.58(0.12–2.72)0.486
AG+GG291.04(0.83–1.31)0.741

a Numbers may not add up to 100% of subjects due to genotyping failure. All samples that did not give a reliable result in the first round of genotyping were resubmitted to up to two additional rounds of genotyping. Data points that were still not filled after this procedure were left blank.

b OR, odds ratio; CI, confidence interval. All analyses were adjusted for age, gender and nationality. Significant associations (p<0.05) are reported in bold.

Table IV

Associations between blood types determined by 6 SNPs in the ABO gene and risk of PDAC.

Table IV

Associations between blood types determined by 6 SNPs in the ABO gene and risk of PDAC.

Blood groupCasesaControlsaORb(95% CI)bP-valuePtrend
O3438701 (ref)--0.895
A5159681.30(1.09–1.54)0.003
 A1/O3176111.25(1.02–1.51)0.028
 A1/A1691121.57(1.11–2.23)0.011
 A1/A232511.71(1.04–2.80)0.033
 A2/O891871.10(0.82–1.49)0.517
 A2/A2270.79(0.16–3.87)0.769
B1202951.04(0.80–1.35)0.764
AB481420.79(0.54–1.14)0.208

a Numbers may not add up to 100% of subjects due to genotyping failure. All samples that did not give a reliable result in the first round of genotyping were resubmitted to up to two additional rounds of genotyping. Data points that were still not filled after this procedure were left blank.

b OR, odds ratio; CI, confidence interval. All analyses were adjusted for age, gender and nationality. Significant associations (p< 0.05) are reported in bold.

In comparison to individuals with O blood type, those with A blood type were at greater risk of developing pancreatic cancer (OR 1.24; 95% CI 1.04–1.48). Individuals with B or AB blood types did not show any increase in risk. To address the hypothesis raised by Wolpin et al(12) that the A1 allele confers a greater risk of pancreatic cancer than the A2 allele, we estimated ORs for A1 and A2 alleles compared with O, which was considered as the reference category. By considering all the study population together we observed that individuals with genotype A2/O had OR 1.14 (95% CI 0.84–1.54), genotype A2/A1 had OR 1.76 (95% CI 1.08–2.89), genotype A1/O had OR 1.28 (95% CI 1.06–1.56) and genotype A1/A1 had OR 1.68 (95% CI 1.14–2.30). Only 2 cases and 7 controls inherited an A2/A2 genotype, severely limiting our ability to accurately assess risk for subjects with this genotype. These analyses are shown in Table IV. When performing subgroup analysis according to country of origin we had comparable results (data not shown).

To address the hypothesis that ABO blood groups may also have prognostic significance, we analyzed the SNPs and the resulting blood groups in relation to patient survival. We found no evidence that genetic variability at this locus modifies the survival of pancreatic cancer, when taking into account multiple testing. We performed stratified analysis considering country of origin and TNM status and we observed no statistically significant association (data not shown). The results of the analysis on the whole consortium are shown in Table V.

Table V

Associations between ABO SNPs and blood types and overall survival of PDAC patients.

Table V

Associations between ABO SNPs and blood types and overall survival of PDAC patients.

SNP or blood groupAnalysisSubjectsaDeathsaHRb95% CIbP-value
rs8176720Per allele9377610.94(0.84–1.05)0.264
AA vs. AG0.91(0.78–1.06)0.240
AA vs. GG0.91(0.72–1.15)0.440
AA vs. (AG+GG)0.91(0.79–1.05)0.213
rs8176741Per allele9227490.82(0.68–0.99)0.036
CC vs. CT0.79(0.64–0.98)0.032
CC vs. TT0.81(0.44–1.52)0.521
CC vs. (CT+TT)0.79(0.65–0.97)0.027
s8176746Per allele9417680.86(0.72–1.03)0.100
CC vs. CA0.86(0.70–1.05)0.139
CC vs. AA0.76(0.39–1.48)0.422
CC vs. (CA+AA)0.85(0.70–1.03)0.105
rs8176747Per allele9447690.85(0.71–1.02)0.077
GG vs. CG0.85(0.69–1.04)0.105
GG vs. GG0.76(0.39–1.47)0.416
GG vs. (CG+GG)0.84(0.69–1.02)0.079
rs505922Per allele9397630.98(0.83–1.15)0.797
TT vs. CT1.06(0.85–1.31)0.610
TT vs. CC1.00(0.86–1.16)0.971
TT vs. (CT+CC)1.02(0.92–1.13)0.724
A9247491.04(0.89–1.22)0.604
B0.86(0.67–1.10)0.234
AB0.90(0.62–1.30)0.577

a Numbers may not add up to 100% of subjects due to genotyping failure. All samples that did not give a reliable result in the first round of genotyping were resubmitted to up to two additional rounds of genotyping. Data points that were still not filled after this procedure were left blank.

b HR, hazard ratio; CI, confidence interval. No association remained statistically significant when multiple testing was taken into account. All analyses were adjusted for age, gender, TNM stage and nationality.

Discussion

Pancreatic cancer is among the most aggressive types of cancer, with mortality rates approaching incidence rates (1,22,23). There is currently no effective curative treatment for pancreatic cancer. Surgery offers the best treatment option and together with some chemotherapeutic treatments it significantly improves survival. Finding genetic variants associated with disease risk is therefore of the utmost importance to define population subgroups who are at high risk of developing the disease. Furthermore, identification of genetic factors associated with progression and survival can significantly contribute to identifying patients with better prognosis and may therefore help clinicians in the choice of treatment options. As there are few known risk factors, improved diagnostics and a better understanding of the molecular pathogenesis of this disease are urgently needed.

In this study, we investigated two endpoints related to ABO glycosyltransferase activity in 1,028 cases and 2,257 controls recruited in the context of the PANDoRA consortium. ABO blood group antigens are found in several tissues and in particularly great abundance on epithelial cells. They represent terminal glycosylation steps of glycoproteins or glycolipid (24). Glycoconjugates, such as the ABO antigen, are important mediators of intercellular adhesion and membrane signaling, which are both critical for the progression and spread of malignant cells (25). Moreover, as cell surface molecules they are also recognized by the host immune response and may influence immunosurveillance for malignant cells (26).

Initially, we evaluated the impact of functional genetic polymorphisms on pancreatic cancer risk. We observed that the minor allele of three SNPs (rs8176741, rs8176746 and rs8176747) was associated with decreased risk of pancreatic cancer while rs505922 was associated with increased risk of developing the disease. The remaining two SNPs did not show any association. Polymorphisms rs8176741, rs8176746 and rs8176747 are in high linkage disequilibrium in our population (mean r2=0.95) and therefore reflect the same association. We typed all of them to have a complete assessment of the phenotypes. By estimating the risk of pancreatic cancer according to the blood type we found that only carriers of the A allele were at increased risk of the disease, while carriers of the B allele were not. This finding is significant considering the fact that Wolpin et al reported an increase of risk for carriers of A or B allele compared to O carriers (1012). This is not due to lack of statistical power in our study; we had more than 87% power to replicate the association with the AB group and more than 99% for the B group. Our study and the study by Wolpin et al have comparable sizes and the frequencies of the ABO alleles are similar in the controls, while they differ slightly in the cases (the frequency of AB was 15% in PanScan and 10% in PANDoRA). The discrepancy in the results could be explained by the fact that PanScan individuals were drawn from prospective cohort studies, while PANDoRA subjects were collected from case control studies. Divergent results in cohort and case control studies have already been reported for pancreatic cancer. An example is the association of Sonic Hedgehog (SHH) polymorphisms, associated with increased pancreatic cancer risk in the PanScan prospective cohorts but not in the PanScan case-control studies (6,8).

Wolpin et al(11) suggested that the association of pancreatic cancer with the A allele appears to be predominantly due to the A1 glycosyltransferase, which exhibits higher activity than A2 glycosyltransferase (13). In our analysis, indeed, the less efficient enzyme A2 did not increase the disease risk in comparison with the non-active O glycosyltransferase.

The results of these two large studies taken together offer a strong direct link between the ABO glycosyltransferase activity and the pathogenesis of pancreatic cancer and suggest that ABO status is not only a marker for pancreatic cancer risk but an actual causative factor for developing the disease. The latter hypothesis remains to be confirmed by functional studies. Since risk factors may also have prognostic significance for pancreatic cancer, as suggested (27), we addressed the hypothesis that ABO blood types and the genetic variability at the locus could be implicated in patient survival. To date, this has been examined only in studies with a smaller sample size than ours (16,17) with inconsistent results. The samples from Germany partially overlap with those used in a recent study (28). Due to the lack of information on CA19-9, lymph node status and grading, we were not able to replicate the statistically significant (0.037) association they found using a multivariate analysis. In the univariate analysis, neither study found any statistically significant effect of ABO on survival.

In conclusion, we have confirmed the association of the A blood group with increased risk of pancreatic cancer and we have shown that the risk is mainly due to the A1 allele, establishing a direct connection between the enzymatic activity and the increased risk.

References

1 

Ferlay J, Shin HR, Bray F, Forman D, Mathers C and Parkin DM: Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer. 127:2893–2917. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Anderson K, Mack T and Silverman D: Cancer of the pancreas. Oxford University Press; New York, NY: 2006

3 

Hassan MM, Bondy ML, Wolff RA, et al: Risk factors for pancreatic cancer: case-control study. Am J Gastroenterol. 102:2696–2707. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Jones S, Hruban RH, Kamiyama M, et al: Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science. 324:2172009. View Article : Google Scholar : PubMed/NCBI

5 

Tischkowitz MD, Sabbaghian N, Hamel N, et al: Analysis of the gene coding for the BRCA2-interacting protein PALB2 in familial and sporadic pancreatic cancer. Gastroenterology. 137:1183–1186. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Amundadottir L, Kraft P, Stolzenberg-Solomon RZ, et al: Genome-wide association study identifies variants in the ABO locus associated with susceptibility to pancreatic cancer. Nat Genet. 41:986–990. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Low SK, Kuchiba A, Zembutsu H, et al: Genome-wide association study of pancreatic cancer in Japanese population. PloS One. 5:e118242010. View Article : Google Scholar : PubMed/NCBI

8 

Petersen GM, Amundadottir L, Fuchs CS, et al: A genome-wide association study identifies pancreatic cancer susceptibility loci on chromosomes 13q22.1, 1q32.1 and 5p15.33. Nat Genet. 42:224–228. 2010. View Article : Google Scholar

9 

Wu C, Miao X, Huang L, et al: Genome-wide association study identifies five loci associated with susceptibility to pancreatic cancer in Chinese populations. Nat Genet. 44:62–66. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Wolpin BM, Chan AT, Hartge P, et al: ABO blood group and the risk of pancreatic cancer. J Natl Cancer Inst. 101:424–431. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Wolpin BM, Kraft P, Gross M, et al: Pancreatic cancer risk and ABO blood group alleles: results from the pancreatic cancer cohort consortium. Cancer Res. 70:1015–1023. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Wolpin BM, Kraft P, Xu M, et al: Variant ABO blood group alleles, secretor status and risk of pancreatic cancer: results from the pancreatic cancer cohort consortium. Cancer Epidemiol Biomarkers Prev. 19:3140–3149. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Moran AP, Gupta A and Joshi L: Sweet-talk: role of host glycosylation in bacterial pathogenesis of the gastrointestinal tract. Gut. 60:1412–1425. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Yip SP: Sequence variation at the human ABO locus. Ann Hum Genet. 66:1–27. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Ben Q, Wang K, Yuan Y and Li Z: Pancreatic cancer incidence and outcome in relation to ABO blood groups among Han Chinese patients: a case-control study. Int J Cancer. 128:1179–1186. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Dandona M, Gao F, Linehan DC and Wang-Gillam A: Re: ABO blood group and the risk of pancreatic cancer. J Natl Cancer Inst. 102:135–137. 2010. View Article : Google Scholar : PubMed/NCBI

17 

Engin H, Bilir C, Ustun H and Gokmen A: ABO blood group and risk of pancreatic cancer in a Turkish population in Western Blacksea region. Asian Pac J Cancer Prev. 13:131–133. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Campa D, Rizzato C, Capurso G, et al: Genetic susceptibility to pancreatic cancer and its functional characterisation: The PANcreatic Disease ReseArch (PANDoRA) consortium. Dig Liver Dis. Nov 30–2012.(Epub ahead of print). View Article : Google Scholar

19 

Rizzato C, Campa D, Giese N, et al: Pancreatic cancer susceptibility loci and their role in survival. PloS One. 6:e279212011. View Article : Google Scholar : PubMed/NCBI

20 

Day N, Oakes S, Luben R, et al: EPIC-Norfolk: study design and characteristics of the cohort. European Prospective Investigation of Cancer. Br J Cancer. 80(Suppl 1): 95–103. 1999.PubMed/NCBI

21 

Davey GK, Spencer EA, Appleby PN, Allen NE, Knox KH and Key TJ: EPIC-Oxford: lifestyle characteristics and nutrient intakes in a cohort of 33 883 meat-eaters and 31 546 non meat-eaters in the UK. Public Health Nutr. 6:259–269. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Ferlay J, Parkin DM and Steliarova-Foucher E: Estimates of cancer incidence and mortality in Europe in 2008. Eur J Cancer. 46:765–781. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Jemal A, Siegel R, Ward E, et al: Cancer statistics, 2008. CA Cancer J Clin. 58:71–96. 2008. View Article : Google Scholar

24 

Hallouin F, Goupille C, le Cabellec M, Bara J and le Pendu J: Expression of A and H blood-group and of CD44 antigens during chemical rat colonic carcinogenesis. Glycoconj J. 14:801–808. 1997. View Article : Google Scholar : PubMed/NCBI

25 

Hakomori S: Antigen structure and genetic basis of histo-blood groups A, B and O: their changes associated with human cancer. Biochim Biophys Acta. 1473:247–266. 1999. View Article : Google Scholar

26 

Hakomori S: Tumor-associated carbohydrate antigens defining tumor malignancy: basis for development of anti-cancer vaccines. Adv Exp Med Biol. 491:369–402. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Li D, Morris JS, Liu J, et al: Body mass index and risk, age of onset and survival in patients with pancreatic cancer. JAMA. 301:2553–2562. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Rahbari NN, Bork U, Hinz U, et al: AB0 blood group and prognosis in patients with pancreatic cancer. BMC Cancer. 12:3192012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

April 2013
Volume 29 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Rizzato C, Campa D, Pezzilli R, Soucek P, Greenhalf W, Capurso G, Talar-Wojnarowska R, Heller A, Jamroziak K, Khaw K, Khaw K, et al: ABO blood groups and pancreatic cancer risk and survival: Results from the PANcreatic Disease ReseArch (PANDoRA) consortium. Oncol Rep 29: 1637-1644, 2013
APA
Rizzato, C., Campa, D., Pezzilli, R., Soucek, P., Greenhalf, W., Capurso, G. ... Canzian, F. (2013). ABO blood groups and pancreatic cancer risk and survival: Results from the PANcreatic Disease ReseArch (PANDoRA) consortium. Oncology Reports, 29, 1637-1644. https://doi.org/10.3892/or.2013.2285
MLA
Rizzato, C., Campa, D., Pezzilli, R., Soucek, P., Greenhalf, W., Capurso, G., Talar-Wojnarowska, R., Heller, A., Jamroziak, K., Khaw, K., Key, T. J., Bambi, F., Landi, S., Mohelnikova-Duchonova, B., Vodickova, L., Büchler, M. W., Bugert, P., Vodicka, P., Neoptolemos, J. P., Werner, J., Hoheisel, J. D., Bauer, A. S., Giese, N., Canzian, F."ABO blood groups and pancreatic cancer risk and survival: Results from the PANcreatic Disease ReseArch (PANDoRA) consortium". Oncology Reports 29.4 (2013): 1637-1644.
Chicago
Rizzato, C., Campa, D., Pezzilli, R., Soucek, P., Greenhalf, W., Capurso, G., Talar-Wojnarowska, R., Heller, A., Jamroziak, K., Khaw, K., Key, T. J., Bambi, F., Landi, S., Mohelnikova-Duchonova, B., Vodickova, L., Büchler, M. W., Bugert, P., Vodicka, P., Neoptolemos, J. P., Werner, J., Hoheisel, J. D., Bauer, A. S., Giese, N., Canzian, F."ABO blood groups and pancreatic cancer risk and survival: Results from the PANcreatic Disease ReseArch (PANDoRA) consortium". Oncology Reports 29, no. 4 (2013): 1637-1644. https://doi.org/10.3892/or.2013.2285