Purpose: Lynch syndrome is a cancer predisposition syndrome which includes colon cancer. It is caused by inherited defects in DNA mismatch repair genes. Sporadic colon cancers are influenced by exogenous hormones (e.g., postmenopausal hormones); we hypothesized that polymorphisms which influence endogenous hormones would therefore modify age at colon cancer onset among Lynch syndrome mutation carriers.

Experimental Design: We genotyped 146 Caucasian Lynch syndrome mutation carriers for a 5′-untranslated region polymorphism in cytochrome P450 17A1 (CYP17; c.−34T→C) and an exon 4 polymorphism in catechol O-methyltransferase (COMT; c.472G→A); 50 mutation carriers had developed colon or rectal cancer at last contact. We used χ2 tests to assess differences in counts. Kaplan-Meier survival curves and Cox proportional hazard models assessed age at onset of colorectal cancer stratified by CYP17 and COMT genotypes.

Results: Homozygous carriers of the CYP17 C allele were diagnosed with colorectal cancer 18 years earlier than homozygous carriers of the T allele. Hazard ratios identified that, relative to homozygous carriers of the T allele (T/T), carriers of one copy (T/C) and two copies (C/C) of the rare allele were, respectively, at 1.9-fold and 2.9-fold increased the risk of colon cancer at any age. The COMT rare allele suggested a nonstatistically significant trend of decreased colon cancer risk.

Conclusions: This study showed that a polymorphism in CYP17 (c.−34T→C) modifies age at onset of Lynch syndrome. Because of the high risk of colorectal cancer among this group, knowledge of the CYP17 genotype is warranted for genetic counseling and risk assessment. Future work should assess polymorphisms associated with steroid hormones in Lynch syndrome mutation carriers.

The incidence and mortality rates for colorectal cancer in the Canadian province of Newfoundland and Labrador rank among the highest in the world (1), partly explained by a high prevalence of familial cancer (2). Lynch syndrome is an autosomal dominant genetic syndrome characterized by early age at colon cancer diagnosis and the common occurrence of tumors in the gastrointestinal, upper urologic, and female reproductive tracts (3). Lynch syndrome is caused by germ line mutations in genes involved with postreplicative nucleotide mismatch repair; ∼90% of the causative mutations occur in MLH1 (located on chromosome 3p21.3) or MSH2 (located on chromosome 2p22-21; ref. 4). Loss of DNA repair ability leads to microsatellite instability in tumor tissue, a hallmark of Lynch syndrome (5).

Penetrance of MSH2 germ line mutations is high but variable. The consistent finding of lower penetrance for colon cancer in women than in men with the same mismatch repair defects (610) suggests that steroidal hormones, such as estrogen or testosterone, might modify risk. Exogenous estrogens (postmenopausal hormones and oral contraceptives) likely reduce the risk of sporadic colon cancer (11, 12). More specific to Lynch syndrome, oral contraceptives and postmenopausal hormones are linked with decreased microsatellite instability colorectal cancer risk (13). Recent work by our group suggests that hormonal contraceptives and postmenopausal hormones are associated with decreased colorectal cancer risk among women from families that meet the Amsterdam or revised Bethesda criteria for Lynch syndrome.5

5

Campbell PT, Newcomb P, Gallinger S, Cotterchio M, McLaughlin JR. Exogenous hormones and colorectal cancer risk in Canada: associations stratified by clinically defined familial risk. Cancer Causes Control. In press 2007.

Common genetic polymorphisms have been reported to influence endogenous hormone levels (14, 15). Polymorphisms in the sex steroid synthesis and metabolism genes cytochrome P450 17A1 (CYP17) and catechol O-methyltransferase (COMT) have been studied extensively in relation to risk of breast cancer (1619), and to a lesser extent, cancers of the ovary (20), endometrium (21), and prostate (22, 23).

CYP17 encodes a protein involved in the production of precursors to testosterone, estrone, and estradiol. A common single base pair substitution (c.−34T→C) in the 5′ untranslated region of CYP17 creates an additional promoter site and increases its transcription (24). Relative to the common allele (T), the rare allele (C) correlates with higher serum levels of various sex steroids (e.g., testosterone, progesterone, estrone, and estradiol; refs. 2528).

COMT encodes an enzyme responsible for metabolizing catechol estrogens into their less active derivatives (29). A G→A transition in exon 4 of COMT creates a valine-to-methionine substitution; the substitution was reported to decrease protein activity and stability by 2- to 3-fold in vitro (30, 31). The rare allele (MET) was associated with higher concentrations of several sex hormones in both premenopausal (32) and postmenopausal women (28).

To our knowledge, only one study has examined the influence of one of these polymorphisms (COMT) with risk of sporadic colorectal cancer (33), and no study has been conducted among confirmed Lynch syndrome mutation carriers. Given the putative evidence for a link between steroidal hormone exposures and colorectal cancer, the current interest was to identify whether polymorphisms in CYP17 and COMT modified Lynch syndrome age at colon cancer onset in a sample of 146 confirmed MSH2 mutation carriers. In addition to the reported influences of these polymorphisms on endogenous steroid levels, they were also selected for study because both rare alleles were reported to be sufficiently common in Caucasian populations (15, 3437).

Over the last 25 years, members of more than 100 nuclear families with extensive colorectal cancers were referred to the Provincial Medical Genetics Program at Memorial University of Newfoundland in St. John's, Newfoundland and Labrador, Canada. Members of more than 50 of these families received MSH2 germ line mutation screening for Lynch syndrome (6). Twelve families harbored a founder mutation in MSH2 (c.942+3A→T). One of these families was used in the original linkage study of the 2p locus (38). A second mutation in MSH2 was recently identified, c.646-?_2802+?del (a genomic deletion of exons 4-16), in a very large kindred (39). A third mutation in MSH2, c.1277-?_1386+?del (a genomic deletion of exon 8), was identified in five families (39).

Genomic DNA was extracted from leukocytes using a common salting out procedure (40). Techniques for MSH2 mutation detection were reported previously (6, 39). For the current study, we selected all archived mutation-positive DNA samples for which we had data on age and site of cancer diagnoses, colon screening history, polyp detection, and vital status (n = 161). From those 161 samples, 15 were excluded because of low DNA yield. Written informed consent was obtained from all subjects for use of their DNA samples for research purposes consistent with this study. The ethics review board of the Memorial University of Newfoundland approved this study.

CYP17 and COMT genotypes were determined by PCR and RFLP techniques as previously described (20), with slight adaptation. A 459 bp fragment of CYP17 was amplified using the following primers: forward 5′-TTCTTCCACAAGGCAAGAGA-3′; reverse 5′-TTGGGCCAAAACAAATAAGC-3′. The 25 μL PCR reaction for CYP17 included 100 ng of genomic DNA, 1× reaction buffer, 3.0 mmol/L of MgCl2, 0.2 mmol/L of each deoxynucleotide triphosphate, 0.04 units/μL of Taq polymerase, and 0.6 μmol/L each of forward and reverse primers. The region containing the COMT exon 4 polymorphism was amplified using the primers: forward 5′-TACTGTGGCTACTCAGCTGTGC-3′; and reverse 5′-GTGAACGTGGTGTGAACACC-3′. The 20 μL PCR reaction for COMT included 200 ng of genomic DNA, 1× reaction buffer, 0.2 mmol/L of each deoxynucleotide triphosphate, 0.05 units/μL of Taq polymerase, and 0.2 μmol/L each of forward and reverse primers.

The thermocycler conditions used were 35 cycles of denaturation for 1 min at 94°C, annealing for 1 min at 57°C, and extension for 1 min at 72°C. Initial denaturation (95°C for 5 min) and final extension (72°C for 10 min) steps were also used. After amplification, 3.3 μL of digestion buffer, 0.66 μL of restriction enzyme MspA1 (New England BioLabs), and 0.33 μL of bovine serum albumin were added to each 25 μL sample of the CYP17 PCR product. Digestion of the total mixture occurred for ∼12 h at 37°C. Added to each 20 μL sample of the COMT PCR product were 3 μL of digestion buffer and 1.5 μL of restriction enzyme NlaIII (New England BioLabs). Digestion of the total mixture occurred for 1 h at 37°C. The digested products were separated using agarose gel electrophoresis and visualized using ethidium bromide staining. Restriction fragments were detected as previously reported (16, 24, 36). The presence of a single 459 bp fragment for CYP17 indicated the homozygous common genotype (T/T); fragments of 459, 335, and 124 bp indicated heterozygosity (T/C); and fragments of 335 and 124 bp indicated the homozygous rare genotype (C/C). For COMT, the presence of the MET allele (rare) was shown by a 96 bp fragment; the VAL allele (common) was indicated by a 114 bp fragment. Genotypes were determined blinded to the subjects' cancer status. All genotyping was repeated by automated sequencing to confirm the original results. Discrepancies between the two techniques occurred in three samples. The sequencing results were taken as the gold standard.

Hardy-Weinberg equilibrium was tested among both genotypes, stratified by outcome, and in the total sample. Age at first colon cancer was the outcome of interest in this study. Kaplan-Meier survival curves were used to assess age at first colon cancer diagnosis, stratified by genotype. Homogeneity between survival curves was assessed with the log-rank test. Censored observations occurred where a subject had no colon or rectal cancer diagnosis at last follow-up, or had been diagnosed with colon polyps upon screening, or had been diagnosed with extracolonic cancer. Censoring was implemented in the latter two situations because the natural history of Lynch syndrome was likely altered. The median age at diagnosis represents the age at which 50% of the sample was colon cancer–free. Cox proportional hazard models were used to estimate hazard ratios (HR) and 95% confidence intervals (95% CI): age at colon cancer onset was the modeled outcome, whereas genotype, sex, and MSH2 mutation type were included as covariates. We selected 92 DNA samples from population-based controls in Newfoundland to compare allele frequencies in the current study with that of the general population. Controls were identified through random-digit dialing and frequency-matched on age and sex to the case population and were unaffected by any cancer. These analyses were conducted to assess potential population stratification in this study. All analyses were conducted with the SAS software package, version 9.1 (SAS Institute).

As shown in Table 1, 50 of the 146 MSH2 mutation carriers developed colon cancer at last follow-up. The majority of the total sample was female (59%). The majority of the colon cancer sample was male (58%). All participants were Caucasian. Mean age at colon cancer diagnosis was slightly younger among males (38 years) than females (43 years), and the difference was statistically significant according to a t test (P = 0.043). Mean age at colon cancer diagnosis and age at censoring were similar between the two outcome groups, as shown in Table 1 (P = 0.89).

Table 1.

Demographic and genetic characteristics of the study sample

MeasureColon cancer, n = 50No colon cancer, n = 96Total, n = 146 (%)*
Sex    
    Male 29 31 60 (41) 
    Female 21 65 86 (59) 
Age at colon cancer diagnosis or censoring    
    Mean (SD) 39.8 (8.9) 39.6 (12.2) 39.6 (11.2) 
    Minimum and maximum 19-63 19-82 19-82 
COMT genotype    
    Val/Val 10 26 36 (25) 
    Val/Met 23 45 68 (46) 
    Met/Met 17 25 42 (29) 
CYP17 genotype    
    T/T 13 42 55 (38) 
    T/C 28 47 75 (51) 
    C/C 16 (11) 
hMSH2 germ line mutation type    
    c.942+3A→T 23 64 87 (60) 
    c.646-?_2802+?del (exon 4-16 del) 14 21 35 (24) 
    c.1277-?_1386+?del (exon 8 del) 13 11 24 (16) 
MeasureColon cancer, n = 50No colon cancer, n = 96Total, n = 146 (%)*
Sex    
    Male 29 31 60 (41) 
    Female 21 65 86 (59) 
Age at colon cancer diagnosis or censoring    
    Mean (SD) 39.8 (8.9) 39.6 (12.2) 39.6 (11.2) 
    Minimum and maximum 19-63 19-82 19-82 
COMT genotype    
    Val/Val 10 26 36 (25) 
    Val/Met 23 45 68 (46) 
    Met/Met 17 25 42 (29) 
CYP17 genotype    
    T/T 13 42 55 (38) 
    T/C 28 47 75 (51) 
    C/C 16 (11) 
hMSH2 germ line mutation type    
    c.942+3A→T 23 64 87 (60) 
    c.646-?_2802+?del (exon 4-16 del) 14 21 35 (24) 
    c.1277-?_1386+?del (exon 8 del) 13 11 24 (16) 
*

Parentheses denote percentages, unless otherwise noted.

COMT and CYP17 genotype frequencies did not differ by sex (COMT: χ2 = 0.49, P = 0.78; CYP17: χ2 = 2.7, P = 0.25). COMT and CYP17 rare alleles were similar in the current study and 92 controls selected from the general population (COMT: χ2 = 2.37, P = 0.31; CYP17: χ2 = 0.015, P = 0.99). Among the entire sample, and in the unaffected group, both COMT and CYP17 genotypes were in Hardy-Weinberg equilibrium. The c.942+3A→T MSH2 mutation was most common in our sample (60%), owing to the greater duration of contact with the Provincial Medical Genetics Program. Colon cancers were proportionately more common among carriers of the c.1277-?_1386+?del mutation (54% affected) than among the c.646-?_2802+?del (40% affected) or c.942+3A→T (26% affected) mutations (χ2 = 7.1, P = 0.03).

Age at colon cancer onset did not differ by COMT genotype in the entire sample or when stratified by sex (Table 2; Fig. 1). HRs and CIs, including variables for sex, and MSH2 allele, were consistent with these findings (Val/Met versus Val/Val HR, 0.89; 95% CI, 0.41-1.91; Met/Met versus Val/Val HR, 0.73; 95% CI, 0.31-1.71).

Table 2.

Kaplan-Meier survival results for age at onset, stratified by COMT and CYP17 genotypes

Median age at onset* (n)
Log-rank χ2P
Homozygous commonHeterozygousHomozygous rare
COMT genotype      
    Total sample 47 (36) 49 (68) 48 (42) 0.034 0.98 
    Males 47 (13) 41 (29) 43 (18) 0.23 0.89 
    Females NA (23) 58 (39) 56 (24) 0.41 0.81 
CYP17 genotype      
    Total sample 63 (55) 48 (75) 45 (16) 6.12 0.047 
    Males 63 (27) 39 (26) 41 (7) 4.27 0.118 
    Females NA (28) 58 (49) 45 (9) 3.31 0.19 
Median age at onset* (n)
Log-rank χ2P
Homozygous commonHeterozygousHomozygous rare
COMT genotype      
    Total sample 47 (36) 49 (68) 48 (42) 0.034 0.98 
    Males 47 (13) 41 (29) 43 (18) 0.23 0.89 
    Females NA (23) 58 (39) 56 (24) 0.41 0.81 
CYP17 genotype      
    Total sample 63 (55) 48 (75) 45 (16) 6.12 0.047 
    Males 63 (27) 39 (26) 41 (7) 4.27 0.118 
    Females NA (28) 58 (49) 45 (9) 3.31 0.19 

Abbreviation: NA, not available (<50% of the category was affected by colon cancer).

*

Median age at onset was defined as the point at which 50% of the samples were unaffected by a first colon cancer diagnosis.

Fig. 1.

COMT genotype and age at colon cancer onset.

Fig. 1.

COMT genotype and age at colon cancer onset.

Close modal

Age at colon cancer onset differed among the entire sample when stratified by CYP17 genotype. In the entire sample, the median age at colon cancer diagnosis among CYP17 T/T, T/C, and C/C subjects was 63, 48, and 45 years, respectively (Table 2; Fig. 2). After stratifying by sex, risk was attenuated toward null in both groups. Among the entire sample, multivariable HRs and 95% CIs were consistent with the Kaplan-Meier analyses (T/C versus T/T HR, 1.89; 95% CI, 0.94-3.75; C/C versus T/T HR, 2.85; 95% CI, 1.13-7.16).

Fig. 2.

CYP17 genotype and age at colon cancer onset.

Fig. 2.

CYP17 genotype and age at colon cancer onset.

Close modal

Our data suggested that a common polymorphism in CYP17 modified age at colon cancer onset among MSH2 germ line mutation carriers. Compared with homozygote carriers of the common allele (T/T), risk of colon cancer was increased by nearly 2-fold among heterozygote carriers (T/C) and risk was further increased by nearly 3-fold among homozygote carriers of the rare allele (C/C). These results suggest a gene-dosage (or additive) effect for the C allele (P for linear trend = 0.02). In our data, a common COMT polymorphism was not associated with age at colon cancer onset; however, homozygous carriers of the rare allele (MET/MET) relative to homozygous carriers of the common allele (VAL/VAL) experienced a nonsignificant 27% reduction in risk of colorectal cancer at any given age.

These findings for CYP17 were consistent with previous reports of the effects of polymorphisms in other genes among Lynch syndrome mutation carriers. For example, clinic-based data from the United States showed that carriers of a variant allele in cyclin D1 developed Lynch syndrome an average of 11 years earlier than noncarriers (41). Similar work from the same group showed that Lynch syndrome age at onset was modified by rare alleles in NAT2 (42), p53 (43), DNMT3b (44), and IGF1 (45). Work in Germany reported that polymorphisms in RNASEL (46) and p53 (47) modified age at onset of Lynch syndrome. Work from South Africa recently reported that variants in GSTT1 and GSTM1 were associated with earlier age at onset of Lynch syndrome among a homogeneous sample of MLH1 mutation carriers (48). We found that COMT did not significantly modify Lynch syndrome age at onset; similarly, others have reported that age at onset of Lynch syndrome was not modified by polymorphisms in ATM (49), GSTM1 (50), CHEK2 (51), p53 (52), and cyclin D1 (53, 54).

To our knowledge, no previous study has examined the effect of the CYP17 or COMT polymorphisms on colon cancer risk among Lynch syndrome mutation carriers. Furthermore, we are not aware of any studies which have evaluated the risk of sporadic colon cancer with the CYP17 genotype. The CYP17 c.−34T→C polymorphism has been examined extensively in breast cancer with earlier studies, suggesting an increased risk of disease from the C allele (16, 17), whereas more recent studies and meta-analyses do not support this link (18, 19). Evidence for a link between the CYP17 c.−34T→C polymorphism and cancers of the ovary (20), endometrium (21), and prostate (22, 23) were similarly inconsistent and controversial. Landi et al. (33) reported no association between the current COMT polymorphism and risk of sporadic colon cancer in a case-control study. Given the relatively small sample size in our study, and the suggestive trend reported herein, future studies of this allele are warranted.

These polymorphisms are plausible candidates for colon cancer studies because of their effects on altered endogenous sex hormone levels. Given the higher levels of endogenous estrone (55), estradiol (25, 55), and progesterone (25, 55) associated with the CYP17 C allele relative to the T allele, combined with the consistent evidence that supplementation with these hormones decreases the risk of colorectal cancer in women (11, 12), our finding of a deleterious effect for the C allele was partly unexpected. This unexpected finding might be explained by pleiotropy: whereas C allele–associated increases in estrogens may decrease risk of colorectal cancer in women, the C allele has also been associated with increased levels of testosterone, androstenedione, and SHBG (28, 5557). Several of these hormones have been implicated in colon tumorigenesis (5863). Supplemental estrogens in males caused insulin resistance and hyperinsulinemia (64); markers of insulin resistance and diabetes are both major risk factors for colorectal cancer (65), supporting the link between the C allele and increased colorectal cancer risk in males. Alternatively, the C allele may be in linkage disequilibrium with another, unknown, polymorphism that confers risk on colon cancer. We found no evidence to support this alternative explanation in the literature or in online repositories of gene sequence data.

The strengths of this study include that it was conducted among participants who largely share common geographic and environmental conditions; this homogeneity should, by design, hold other potentially confounding issues to a minimum and likely decrease the risk of population stratification. Additionally, all subjects in this study shared deleterious defects in the same gene. Although the environmental and genetic homogeneity strengthens the internal validity of this study, the external validity is weakened, further highlighting the need for future studies in other populations. Frequencies of the rare alleles (COMT, 52%; CYP17, 37%) in this study were similar to previous reports among Caucasian populations in the United Kingdom, United States, and Canada (15, 3437, 6671), strengthening the external validity in this study. Furthermore, the frequencies of the rare alleles in the current study were similar to a randomly selected sample of 92 population-based controls from Newfoundland (COMT, 48%; CYP17, 37%), supporting the fact that population stratification did not account for these results. Ideally, future studies would also collect data on lifestyle and demographic data; it was not possible for us to collect this information.

One potential bias in this study was from the reported effect of the C allele on postmenopausal hormone use; female carriers with the C/C relative to the T/T genotype were previously reported to be about half as likely to use postmenopausal hormones (72). Although we do not have data on postmenopausal hormone use, we feel this potential for bias was likely minimal. The women in this cohort were relatively young and colon cancers and censoring occurred at relatively young ages (mean age at colon cancer diagnosis among females, 42.8 years; mean age at censoring among females, 40.2 years). All but 3 of the 21 female colon cancers occurred prior to age 50 years. Postmenopausal hormones were largely irrelevant in this sample because their cancers occurred, on average, about one decade prior to menopause. Of the censored women, 10 of 65 were older than 50 years. In the absence of questionnaire or pharmacy record data, we cannot exclude the possibility that some of them used postmenopausal hormones, and we cannot rule out that postmenopausal hormone use might have differed according to CYP17 genotype. Although anecdotal evidence from clinic staff suggests that exogenous hormone use in this population is very low.

Given the dramatic increased lifetime risk of colon and extracolonic cancers among germ line mutation carriers linked with Lynch syndrome, knowledge of genetic and environmental modifiers is needed for risk counseling and targeted prevention strategies. This study found that a common allele in CYP17 modified age at colon cancer onset among MSH2 mutation carriers. Future studies are needed to confirm this association; examination of other polymorphisms involved with steroid production and metabolism is also warranted.

Grant support: Canadian Institutes of Health Research (CRT 43821) and the Newfoundland Cancer Treatment and Research Foundation. National Cancer Institute of Canada Doctoral Fellowship (no. 13523), with funds from the Canadian Cancer Society (P.T. Campbell). National Cancer Institute of Canada Post-Doctoral Fellowship (no. 13493), with funds from the Canadian Cancer Society (M.O. Woods).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1
Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002.
CA Cancer J Clin
2005
;
55
:
74
–108.
2
Green RC, Buehler S, Gallinger S, et al. Very high incidence of familial colorectal cancer in Newfoundland. A comparison with 13 other population-based registries. Fam Cancer 2006; In press.
3
Abdel-Rahman WM, Mecklin JP, Peltomaki P. The genetics of HNPCC: application to diagnosis and screening.
Crit Rev Oncol Hematol
2006
;
58
:
208
–20.
4
Peltomaki P. Lynch syndrome genes.
Fam Cancer
2005
;
4
:
227
–32.
5
Baudhuin LM, Burgart LJ, Leontovich O, Thibodeau SN. Use of microsatellite instability and immunohistochemistry testing for the identification of individuals at risk for Lynch syndrome.
Fam Cancer
2005
;
4
:
255
–65.
6
Green J, O'Driscoll M, Barnes A, et al. Impact of gender and parent of origin on the phenotypic expression of hereditary nonpolyposis colorectal cancer in a large Newfoundland kindred with a common MSH2 mutation.
Dis Colon Rectum
2002
;
45
:
1223
–32.
7
Dunlop MG, Farrington SM, Carothers AD, et al. Cancer risk associated with germline DNA mismatch repair gene mutations.
Hum Mol Genet
1997
;
6
:
105
–10.
8
Froggatt NJ, Green J, Brassett C, et al. A common MSH2 mutation in English and North American HNPCC families: origin, phenotypic expression, and sex specific differences in colorectal cancer.
J Med Genet
1999
;
36
:
97
–102.
9
Vasen HF, Wijnen JT, Menko FH, et al. Cancer risk in families with hereditary nonpolyposis colorectal cancer diagnosed by mutation analysis.
Gastroenterology
1996
;
110
:
1020
–7.
10
Jenkins MA, Baglietto L, Dowty JG, et al. Cancer risks for mismatch repair gene mutation carriers: a population-based early onset case-family study.
Clin Gastroenterol Hepatol
2006
;
4
:
489
–98.
11
Fernandez E, La Vecchia C, Balducci A, Chatenoud L, Franceschi S, Negri E. Oral contraceptives and colorectal cancer risk: a meta-analysis.
Br J Cancer
2001
;
84
:
722
–7.
12
La Vecchia C, Gallus S, Fernandez E. Hormone replacement therapy and colorectal cancer: an update.
J Br Menopause Soc
2005
;
11
:
166
–72.
13
Slattery ML, Potter JD, Curtin K, et al. Estrogens reduce and withdrawal of estrogens increase risk of microsatellite instability-positive colon cancer.
Cancer Res
2001
;
61
:
126
–30.
14
Platz EA, Giovannucci E. The epidemiology of sex steroid hormones and their signaling and metabolic pathways in the etiology of prostate cancer.
J Steroid Biochem Mol Biol
2004
;
92
:
237
–53.
15
Sharp L, Cardy AH, Cotton SC, Little J. CYP17 gene polymorphisms: prevalence and associations with hormone levels and related factors. a HuGE review.
Am J Epidemiol
2004
;
160
:
729
–40.
16
Feigelson HS, Coetzee GA, Kolonel LN, Ross RK, Henderson BE. A polymorphism in the CYP17 gene increases the risk of breast cancer.
Cancer Res
1997
;
57
:
1063
–5.
17
Bergman-Jungestrom M, Gentile M, Lundin AC, Wingren S. Association between CYP17 gene polymorphism and risk of breast cancer in young women.
Int J Cancer
1999
;
84
:
350
–3.
18
Little J, Simard J. CYP17 and breast cancer: no overall effect, but what about interactions?
Breast Cancer Res
2005
;
7
:
238
–42.
19
Chang JH, Gertig DM, Chen X, et al. CYP17 genetic polymorphism, breast cancer, and breast cancer risk factors: Australian Breast Cancer Family Study.
Breast Cancer Res
2005
;
7
:
R513
–21.
20
Garner EI, Stokes EE, Berkowitz RS, Mok SC, Cramer DW. Polymorphisms of the estrogen-metabolizing genes CYP17 and catechol-O-methyltransferase and risk of epithelial ovarian cancer.
Cancer Res
2002
;
62
:
3058
–62.
21
Szyllo K, Smolarz B, Romanowicz-Makowska H, Kulig A. The polymorphisms of the CYP17 and CYP19 genes in endometrial cancer patients.
Pol J Pathol
2006
;
57
:
35
–40.
22
Sobti RC, Onsory K, Al-Badran AI, et al. CYP17, SRD5A2, CYP1B1, and CYP2D6 gene polymorphisms with prostate cancer risk in North Indian population.
DNA Cell Biol
2006
;
25
:
287
–94.
23
Vesovic Z, Herkommer K, Vogel W, Paiss T, Maier C. Role of a CYP17 promoter polymorphism for familial prostate cancer risk in Germany.
Anticancer Res
2005
;
25
:
1303
–7.
24
Carey AH, Waterworth D, Patel K, et al. Polycystic ovaries and premature male pattern baldness are associated with one allele of the steroid metabolism gene CYP17.
Hum Mol Genet
1994
;
3
:
1873
–6.
25
Feigelson HS, Shames LS, Pike MC, Coetzee GA, Stanczyk FZ, Henderson BE. Cytochrome P450c17α gene (CYP17) polymorphism is associated with serum estrogen and progesterone concentrations.
Cancer Res
1998
;
58
:
585
–7.
26
Daneshmand S, Weitsman SR, Navab A, Jakimiuk AJ, Magoffin DA. Overexpression of theca-cell messenger RNA in polycystic ovary syndrome does not correlate with polymorphisms in the cholesterol side-chain cleavage and 17α-hydroxylase/C(17–20) lyase promoters.
Fertil Steril
2002
;
77
:
274
–80.
27
Haiman CA, Hankinson SE, Spiegelman D, et al. The relationship between a polymorphism in CYP17 with plasma hormone levels and breast cancer.
Cancer Res
1999
;
59
:
1015
–20.
28
Tworoger SS, Chubak J, Aiello EJ, et al. Association of CYP17, CYP19, CYP1B1, and COMT polymorphisms with serum and urinary sex hormone concentrations in postmenopausal women.
Cancer Epidemiol Biomarkers Prev
2004
;
13
:
94
–101.
29
Zhu BT. Catechol-O-Methyltransferase (COMT)-mediated methylation metabolism of endogenous bioactive catechols and modulation by endobiotics and xenobiotics: importance in pathophysiology and pathogenesis.
Curr Drug Metab
2002
;
3
:
321
–49.
30
Dawling S, Roodi N, Mernaugh RL, Wang X, Parl FF. Catechol-O-methyltransferase (COMT)-mediated metabolism of catechol estrogens: comparison of wild-type and variant COMT isoforms.
Cancer Res
2001
;
61
:
6716
–22.
31
Lotta T, Vidgren J, Tilgmann C, et al. Kinetics of human soluble and membrane-bound catechol O-methyltransferase: a revised mechanism and description of the thermolabile variant of the enzyme.
Biochemistry
1995
;
34
:
4202
–10.
32
Hong CC, Thompson HJ, Jiang C, et al. Val158Met polymorphism in catechol-O-methyltransferase gene associated with risk factors for breast cancer.
Cancer Epidemiol Biomarkers Prev
2003
;
12
:
838
–47.
33
Landi S, Gemignani F, Moreno V, et al. A comprehensive analysis of phase I and phase II metabolism gene polymorphisms and risk of colorectal cancer.
Pharmacogenet Genomics
2005
;
15
:
535
–46.
34
Allen NE, Forrest MS, Key TJ. The association between polymorphisms in the CYP17 and 5α-reductase (SRD5A2) genes and serum androgen concentrations in men.
Cancer Epidemiol Biomarkers Prev
2001
;
10
:
185
–9.
35
Jernstrom H, Vesprini D, Bradlow HL, Narod SA. Re: CYP17 promoter polymorphism and breast cancer in Australian women under age forty years.
J Natl Cancer Inst
2001
;
93
:
554
–5.
36
Lavigne JA, Helzlsouer KJ, Huang HY, et al. An association between the allele coding for a low activity variant of catechol-O-methyltransferase and the risk for breast cancer.
Cancer Res
1997
;
57
:
5493
–7.
37
Thompson PA, Shields PG, Freudenheim JL, et al. Genetic polymorphisms in catechol-O-methyltransferase, menopausal status, and breast cancer risk.
Cancer Res
1998
;
58
:
2107
–10.
38
Peltomaki P, Aaltonen LA, Sistonen P, et al. Genetic mapping of a locus predisposing to human colorectal cancer.
Science
1993
;
260
:
810
–2.
39
Woods MO, Hyde AJ, Curtis FK, et al. High frequency of hereditary colorectal cancer in Newfoundland likely involves novel susceptibility genes.
Clin Cancer Res
2005
;
11
:
6853
–61.
40
Miller SA, Dykes DD, Polesky HF. A simple salting out procedure for extracting DNA from human nucleated cells.
Nucleic Acids Res
1988
;
16
:
1215
.
41
Kong S, Amos CI, Luthra R, Lynch PM, Levin B, Frazier ML. Effects of cyclin D1 polymorphism on age of onset of hereditary nonpolyposis colorectal cancer.
Cancer Res
2000
;
60
:
249
–52.
42
Frazier ML, O'Donnell FT, Kong S, et al. Age-associated risk of cancer among individuals with N-acetyltransferase 2 (NAT2) mutations and mutations in DNA mismatch repair genes.
Cancer Res
2001
;
61
:
1269
–71.
43
Jones JS, Chi X, Gu X, Lynch PM, Amos CI, Frazier ML. p53 polymorphism and age of onset of hereditary nonpolyposis colorectal cancer in a Caucasian population.
Clin Cancer Res
2004
;
10
:
5845
–9.
44
Jones JS, Amos CI, Pande M, et al. DNMT3b polymorphism and hereditary nonpolyposis colorectal cancer age of onset.
Cancer Epidemiol Biomarkers Prev
2006
;
15
:
886
–91.
45
Zecevic M, Amos CI, Gu X, et al. IGF1 gene polymorphism and risk for hereditary nonpolyposis colorectal cancer.
J Natl Cancer Inst
2006
;
98
:
139
–43.
46
Kruger S, Silber AS, Engel C, et al. Arg462Gln sequence variation in the prostate-cancer-susceptibility gene RNASEL and age of onset of hereditary non-polyposis colorectal cancer: a case-control study.
Lancet Oncol
2005
;
6
:
566
–72.
47
Kruger S, Bier A, Engel C, et al. The p53 codon 72 variation is associated with the age of onset of hereditary non-polyposis colorectal cancer (HNPCC).
J Med Genet
2005
;
42
:
769
–73.
48
Felix R, Bodmer W, Fearnhead NS, van der Merwe L, Goldberg P, Ramesar RS. GSTM1 and GSTT1 polymorphisms as modifiers of age at diagnosis of hereditary nonpolyposis colorectal cancer (HNPCC) in a homogeneous cohort of individuals carrying a single predisposing mutation.
Mutat Res
2006
;
602
:
175
–81.
49
Jones JS, Gu X, Lynch PM, Rodriguez-Bigas M, Amos CI, Frazier ML. ATM polymorphism and hereditary nonpolyposis colorectal cancer (HNPCC) age of onset (United States).
Cancer Causes Control
2005
;
16
:
749
–53.
50
Jones JS, Gu X, Campos IM, Lynch PM, Amos CI, Frazier ML. GSTM1 polymorphism does not affect hereditary nonpolyposis colorectal cancer age of onset.
Cancer Epidemiol Biomarkers Prev
2004
;
13
:
676
–8.
51
Sanchez de Abajo A, de la Hoya M, Godino J, et al. The CHEK2 1100delC allele is not relevant for risk assessment in HNPCC and HBCC Spanish families.
Fam Cancer
2005
;
4
:
183
–6.
52
Sotamaa K, Liyanarachchi S, Mecklin JP, et al. p53 codon 72 and MDM2 SNP309 polymorphisms and age of colorectal cancer onset in Lynch syndrome.
Clin Cancer Res
2005
;
11
:
6840
–4.
53
Bala S, Peltomaki P. CYCLIN D1 as a genetic modifier in hereditary nonpolyposis colorectal cancer.
Cancer Res
2001
;
61
:
6042
–5.
54
Kruger S, Engel C, Bier A, et al. Absence of association between cyclin D1 (CCND1) G870A polymorphism and age of onset in hereditary nonpolyposis colorectal cancer.
Cancer Lett
2006
;
236
:
191
–7.
55
Haiman CA, Hankinson SE, Colditz GA, Hunter DJ, De Vivo I. A polymorphism in CYP17 and endometrial cancer risk.
Cancer Res
2001
;
61
:
3955
–60.
56
Onland-Moret NC, van Gils CH, Roest M, Grobbee DE, Peeters PH. Cyp17, urinary sex steroid levels and breast cancer risk in postmenopausal women.
Cancer Epidemiol Biomarkers Prev
2005
;
14
:
815
–20.
57
Hong CC, Thompson HJ, Jiang C, et al. Association between the T27C polymorphism in the cytochrome P450 c17α (CYP17) gene and risk factors for breast cancer.
Breast Cancer Res Treat
2004
;
88
:
217
–30.
58
Izbicki JR, Hamilton SR, Wambach G, et al. Effects of androgen manipulations on chemically induced colonic tumours and on macroscopically normal colonic mucosa in male Sprague-Dawley rats.
Br J Cancer
1990
;
61
:
235
–40.
59
Cintron JR, Asadi FK, Malakouti S, Abcarian HA. Androgen induces ornithine decarboxylase gene expression in colonic cell line HT-29.
Dis Colon Rectum
1996
;
39
:
406
–9.
60
Simard J, Gingras S. Crucial role of cytokines in sex steroid formation in normal and tumoral tissues.
Mol Cell Endocrinol
2001
;
171
:
25
–40.
61
Baris D, Gridley G, Ron E, et al. Acromegaly and cancer risk: a cohort study in Sweden and Denmark.
Cancer Causes Control
2002
;
13
:
395
–400.
62
Berta L, Fronticelli Baldelli C, Fazzari A, et al. Sex steroid receptors, secondary bile acids and colorectal cancer. A possible mechanism of interaction.
Panminerva Med
2003
;
45
:
261
–6.
63
Mestron A, Webb SM, Astorga R, et al. Epidemiology, clinical characteristics, outcome, morbidity and mortality in acromegaly based on the Spanish Acromegaly Registry (Registro Espanol de Acromegalia, REA).
Eur J Endocrinol
2004
;
151
:
439
–46.
64
Polderman KH, Gooren LJ, Asscheman H, Bakker A, Heine RJ. Induction of insulin resistance by androgens and estrogens.
J Clin Endocrinol Metab
1994
;
79
:
265
–71.
65
Potter JD. Colorectal cancer: molecules and populations.
J Natl Cancer Inst
1999
;
91
:
916
–32.
66
Techatraisak K, Conway GS, Rumsby G. Frequency of a polymorphism in the regulatory region of the 17α-hydroxylase-17,20-lyase (CYP17) gene in hyperandrogenic states.
Clin Endocrinol (Oxf)
1997
;
46
:
131
–4.
67
Dunning AM, Healey CS, Pharoah PD, et al. No association between a polymorphism in the steroid metabolism gene CYP17 and risk of breast cancer.
Br J Cancer
1998
;
77
:
2045
–7.
68
Young IE, Kurian KM, Annink C, et al. A polymorphism in the CYP17 gene is associated with male breast cancer.
Br J Cancer
1999
;
81
:
141
–3.
69
Lai J, Vesprini D, Chu W, Jernstrom H, Narod SA. CYP gene polymorphisms and early menarche.
Mol Genet Metab
2001
;
74
:
449
–57.
70
Millikan RC, Pittman GS, Tse CK, et al. Catechol-O-methyltransferase and breast cancer risk.
Carcinogenesis
1998
;
19
:
1943
–7.
71
Miyoshi Y, Noguchi S. Polymorphisms of estrogen synthesizing and metabolizing genes and breast cancer risk in Japanese women.
Biomed Pharmacother
2003
;
57
:
471
–81.
72
Feigelson HS, McKean-Cowdin R, Pike MC, et al. Cytochrome P450c17α gene (CYP17) polymorphism predicts use of hormone replacement therapy.
Cancer Res
1999
;
59
:
3908
–10.