Abstract
Prior epidemiologic findings for plasma folate and B-vitamins and breast cancer risk are inconsistent and have not assessed the influence of folic acid fortification. Therefore, we examined the associations of plasma folate, B12, pyridoxal 5′-phosphate (PLP), homocysteine, cysteine and cysteinylglycine with breast cancer risk, before and after fortification. We conducted a nested case–control study within the prospective Nurses’ Health Study. In 1989–1990 (pre-fortification), 32,826 women donated a blood sample and 18,743 donated an additional blood sample in 2000–2001 (post-fortification). Between the first blood collection and 2006, 1874 incident breast cancer cases with at least one blood sample and 367 with two were 1:1 matched to controls. Conditional logistic regression was used to estimate relative risks (RR) and 95% confidence intervals (CI) adjusting for breast cancer risk factors. Overall, higher plasma folate, B12, PLP, homocysteine, cysteine and cysteinylglycine levels were not associated with breast cancer risk. Associations did not vary by in situ/invasive, hormone receptor status, or tumor molecular subtype. Additionally, associations were null before and after fortification. For example, the RR (95% CI) for the highest versus lowest tertile of 1990 (pre-fortification) plasma folate with 1990–2000 follow-up was 0.93 (0.75–1.16) and for the 2000 plasma folate (post-fortification) with 2000–2006 follow-up the RR (95% CI) was 1.17 (0.79–1.74). Plasma folate, B12, PLP, homocysteine, cysteine and cysteinylglycine were not significantly associated with breast cancer overall, before and after fortification, or with specific tumor molecular subtypes. However, long term associations (>8 years) after the implementation of fortification could not be examined.
Keywords: breast cancer, plasma Folate, plasma B-vitamins, molecular subtypes, folic acid fortification
Introduction
Folate (B9), cobalamin (B12) and B6 are essential, water-soluble vitamins found naturally in green leafy vegetables, legumes and organ meats like liver.1,2 The term folate also includes the synthetic form, folic acid, found in supplements and fortified foods. The United States has mandated that enriched grain products be fortified with folic acid since 1998 to prevent neural tube defects in newborns.3 This has increased average total intakes of folate by approximately 200 to 400 μg/day and led to a 50–100% increase in plasma levels.1 While fortification has reduced neural tube defects and has had other beneficial outcomes (e.g., reduced preterm birth, stroke),2 the potential for adverse effects, such as higher cancer risk, has led to concern.1,4–6
Folate is involved in one-carbon metabolism, an important metabolic cycle that regulates DNA methylation and synthesis.1,7–9 Folic acid is reduced to tetrahydrofolate [THF], converted to 5,10-methylene-THF by a B6-dependent enzyme and then reduced to 5-methyl-THF.9,10 An enzyme dependent on B12 transfers the methyl group to homocysteine, which is converted through a series of steps to S-adenosyl methionine (SAM).8,9 5,10-methylene-THF is important for nucleic acid synthesis, and SAM is the primary methyl group donor for DNA methylation.9 Thus, excess folate might promote neoplastic lesions through DNA synthesis and methylation and inactivation of tumor suppressor genes.1
However, folate and other B-vitamins deficiencies may stimulate carcinogenesis through this same cycle. Folate deficiency results in high homocysteine levels and may lead to cancer through reduced synthesis of SAM resulting in DNA hypomethylation11 and/or increased DNA strand breaks and mutations due to impaired DNA repair systems.1,11–14 B12 deficiency reduces conversion of 5-methyl-THF and therefore lower SAM.7–9 B6 deficiency reduces levels of 5,10-methylene-THF, resulting in increased DNA strand breaks and reduced levels of 5-methyl-THF resulting in lower SAM.15
Several aberrant methylation patterns have been noted in breast tumors including widespread DNA hypomethylation and hypermethylation of tumor suppressor genes suggesting that one-carbon metabolism may play a role in breast cancer carcinogenesis.10 With concern for carcinogenesis at both low and high folate levels, folic acid fortification has been controversial.1,6,16,17 To our knowledge, no studies have assessed plasma levels of folate and other B-vitamins both before and after fortification in relation to breast cancer risk. Additionally, few studies have assessed folate levels in relation to molecular subtypes of breast cancer, with most assessing only estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor 2 (HER2) separately.18,19 Therefore, we examined the associations of plasma folate, B12, pyridoxal 5′-phosphate (PLP; the principal active form of B6), homocysteine, cysteine and cysteinylglycine with breast cancer, including an assessment of nutrient levels before and after fortification in relation to breast cancer subtypes within a nested case–control in the prospective Nurses’ Health Study (NHS).
Materials and Methods
Study population
The NHS is a prospective cohort of 121, 700 female registered nurses aged 30–55 years enrolled in 1976 by completing a mailed baseline questionnaire.20–22 Biennially, participants complete mailed questionnaires updating health information, including most known breast cancer risk factors and diagnoses. In 1989–1990 (pre-fortification), 32,826 women donated a blood sample.23,24 The women arranged to have their blood drawn and shipped back overnight with an ice pack, where it was then processed, aliquoted into plasma, red blood cell and white blood cell components, and stored in continuously monitored liquid nitrogen freezers (<−130 °C). A second blood sample from a subset of these women (n = 18,743) was donated in 2000–2001 (post-fortification) using similar protocols.25 Of the 32,826 women who provided at least one blood sample, the follow-up rate in 2006 was 99%. The study was approved by the Committee on the Use of Human Subjects in Research at the Brigham and Women’s Hospital, Boston, Massachusetts; the completion of the self-administered questionnaire and blood collection was considered to imply informed consent.
Case and control selection
Incident cases of breast cancer were self-reported on the biennial questionnaires and confirmed by medical record review. From first blood collection through 2006, there were 1931 incident breast cancer cases (357 carcinoma in situ, 1,544 invasive) among the women without a history of cancer (except non-melanoma skin) that provided at least one blood sample. We excluded 57 case–control pairs with missing information on plasma measures in one member of the pair, resulting in 1874 case–control pairs included in this analysis, of which 367 case–control pairs had two blood samples. The average time from the 1990 blood collection to breast cancer diagnosis was 97 months (range = 1–202) and 28 months (range = 1–73) from the 2000 blood collection to breast cancer diagnosis among those that donated a second blood. Each breast cancer case was matched to one control without a history of cancer, by year of birth (±1 year), time of blood draw (±2 h), fasting status (≥10 h vs. <10 h), month of blood draw (±1 month), use of postmenopausal hormones within 3 months before blood collection, and menopausal status at blood collection and diagnosis.
Laboratory assays
Plasma concentrations of folate, B12, PLP, total homocysteine, total cysteine and cysteinylglycine were assayed at the Jean Mayer United States Department of Agriculture Human Nutrition Research Center on Aging at Tufts University. Folate and vitamin B12 were measured using a radioassay kit (Bio-Rad, Richmond, CA) or microbial assay for folate after 2000.26 For microbial assays, folate and B12 values above the upper limit of quantification, >63 ng/mL for folate (n = 9 for 1st collection; n = 37 for 2nd collection) and > 2014 pg/mL for B12 (n = 19 for 1st collection; n = 21 for 2nd collection), were assigned the values 63 and 2014 respectively. Plasma levels of the active form of B6, pyridoxal 5′-phosphate (PLP), were determined by an enzymatic procedure using radioactive tyrosine and the apo-enzyme tyrosine.27 Total homocysteine, cysteine and cysteinylglycine were measured using high performance liquid chromatography with fluorescence.28 Cases and controls within matched pairs were assayed in the same analytical run. The laboratory was blinded to case and control status and the order of the specimens in each pair was randomized. Additionally, for case–control pairs diagnosed after 2000 with two blood samples, both blood samples were assayed together in the same analytical run. Assays were conducted in four batches, and quality control samples (~10% of samples) were randomly interspersed to assess laboratory precision. Average within-batch coefficients of variation were less than 10%, except one batch each for folate (12.6%), PLP (13.9%), B12 (12.6%) and homocysteine (13.2%). Outliers were detected and removed using the extreme Studentized deviate many-outlier procedure29 (range of n outliers excluded across plasma measures: n = 2–12 for 1st collection; n = 1–3 for 2nd collection).
Plasma values for folate and B12 differed between 1990 and 2000 blood samples for case–control pairs with two blood samples consistent with fortification and increasing use of supplements. Among the 1990 blood samples, there was evidence of some batch variation. Therefore, for folate and B12 we recalibrated all 1990 blood samples, but not the 2000 blood samples, to an average standard batch for each plasma measure.30 For PLP, homocysteine and cysteinylglycine, since there was no change in the plasma values between 1990 and 2000 blood collection, all batches from 1990 and 2000 blood samples were recalibrated to an average standard batch. Lastly, since the distribution of the fourth batch of cysteine (includes both 1990 and 2000 blood samples) had a lower and narrower range of values than earlier batches, but did not differ between 1990 and 2000 blood samples we recalibrated the three earlier batches to an average standard batch. Using the recalibrated data, we defined quantiles based on the control values for each plasma measure. For cysteine, we created batch specific quantiles for the average of the three earlier batches and for batch four. Secondary analyses excluding the fourth batch of cysteine data did not change conclusions.
Covariate data
Dietary intakes of B-vitamins and alcohol were obtained from food frequency questionnaires (FFQ) administered every four years,31 using the 1990 FFQ for 1990 blood collection and the 2002 FFQ for 2000 blood collection. Information on multivitamin and supplement use was obtained from the 1990 and 2000 biennial questionnaires. Information on breast cancer risk factors were collected from the biennial questionnaires and the questionnaires administered at the time of blood collection; age at menarche, height and body mass index (BMI) at age 18 were collected on the baseline questionnaire in 1976, parity and age at first birth, family history of breast cancer and history of benign breast disease were updated to 1990 or 2000, and current weight and menopausal status were from the 1990 or 2000 questionnaires. Missing values were filled in using the prior questionnaire or by missing indicators (i.e., parity/age at first birth, weight change from age 18) or using the median of all participants (all other covariates). In multivariate models we adjusted for age at menarche, parity/age at first birth, age at menopause if postmenopausal at blood collection, family history of breast cancer in mother or a sister, history of benign breast disease, height, BMI at age 18, weight change since age 18, and alcohol intake at blood collection. Results were essentially unchanged if a menopause status and BMI interaction term was added to our models and hence models with only main effects for these two covariates are presented here.
Tumor/outcome data
Information on tumor invasiveness, grade and hormone receptor status were available from pathology reports. Described previously,32,33 archival formalin-fixed paraffin-embedded breast cancer tissue blocks were obtained for approximately 70% of incident primary breast cancer cases through 2006 and used to construct tissue microarrays. Immunohistochemical staining was performed for estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor 2 (HER2), cytokeratin 5/6 (CK5/6) and epidermal growth factor (EGFR) markers. Luminal A cases were defined as ER-positive and/or PR-positive and HER2-negative and histologic grade 1 or 2. Cases were defined as luminal B if they were either 1) ER-positive and/or PR-positive and HER2-positive or 2) ER-positive and/or PR-positive and HER2-negative and grade 3. HER2-type cases were ER-negative, PR-negative and HER2-positive and basal like cases were ER-negative, PR-negative, HER2-negative and either CK5/6-positive or EGFR-positive.
Statistical analyses
We calculated intraclass correlation coefficients (ICC) and Spearman correlations between 1990 and 2000 blood samples for women with both blood samples. We compared characteristics between cases and controls at both blood collections using the Mantel–Haenszel tests for categorical and paired t-tests for continuous characteristics. Additionally, we examined the correlation between the plasma measures and dietary intakes of the B-vitamins for each blood sample using Spearman correlations among the controls with two blood samples.
For the main analysis using the 1990 blood and follow-up from 1990 to 2006, we calculated relative risks (RR) of incident breast cancer and 95% confidence intervals (CI) for each quintile of plasma measure compared to the lowest quintile using multivariable conditional logistic regression. To test for trend across quintiles, we modeled the median quintile values as a continuous variable and report the Wald P-value. As prior studies have suggested non-linear relationships between B-vitamins and breast cancer, we additionally tested for nonlinearity using cubic splines.
We also assessed in situ, invasive and hormone receptor subtype using quintiles and conditional logistic regression. Due to smaller numbers, we assessed molecular subtype and examined relationship by time point using tertiles and unconditional logistic regression adjusting for matching factors. Tests for subtype heterogeneity were calculated using the methods of Wang et al.34 We also examined associations between plasma measures and breast cancer using plasma measures as a continuous variable [per standard deviation (SD) of natural log—transformed plasma measure].
To assess if associations varied before and after fortification, plasma levels were evaluated as 1) 1990 blood levels with cases diagnosed between 1990 and 2000, 2) 1990 blood levels with cases diagnosed between 2000 and 2006, 3) 2000 blood levels with cases diagnosed between 2000 and 2006. Additionally, to evaluate the 1990 and 2000 plasma measures in the same model, we conducted two analyses. First, we averaged the natural log transformed 1990 and 2000 plasma measures among those with both blood donations. Using the averaged values, in tertile categories and as a 1SD increase continuous value, we used unconditional logistic regression adjusting for the matching factors and breast cancer risk factors at both blood collections to estimate RR and 95% CIs. Second, we conducted a repeated measures logistic regression using PROC GENMOD in SAS, where the 1990 plasma measure was used for the 1990–2000 time period and the 2000 plasma measure was used for the 2000–2006 time period, updating covariates when relevant. Controls from the 2000–2006 case–control pairs were allowed to contribute to both the 1990–2000 and 2000–2006 time periods, but with their 1990 plasma measure used for the 1990–2000 time period and their 2000 plasma measure used for the 2000–2006 time period.
To assess if associations varied across other exposures, we conducted stratified analysis using unconditional logistic regression and to obtain a test of significance we used the multiplicative interaction term of the stratifying variable and continuous plasma measure (per SD of natural log–transformed plasma measure) in the model, and tested via a likelihood ratio test. Secondarily, we also restricted analyses to nonmultivitamin users.
All P-values were based on two-sided tests and were considered statistically significant if p < 0.05. Analyses were conducted using SAS 9.3 (SAS Institute Inc., Cary, NC, USA) for UNIX.
Results
At the first blood collection in 1990, the mean age of the participants was 56.8 years (range = 42.5–70.2) and in 2000, the mean age of the participants was 67.0 years (range = 53.9–80.3). Characteristics by breast cancer case and control status at both blood collections are shown in Table 1. Cases were more likely to have a family history of breast cancer and a history of benign breast disease at the 1990 blood collection. Mean plasma levels at the 2000 blood collection (post-fortification) showed substantial increases in the folate concentrations and smaller increases in B12 and PLP concentrations. Multivitamin use also increased between the two bloods (39% in 1990 and 59% in 2000 among controls).
Table 1.
Characteristics | At the 1990 blood collection | At the 2000 blood collection | ||||
---|---|---|---|---|---|---|
Breast cancer cases (n = 1874) | Controls (n = 1874) | p Value1 | Breast cancer cases (n = 367) | Controls (n = 367) | p Value1 | |
Mean (SD) | Mean (SD) | |||||
Age at blood collection, y | 56.7 (7.0) | 56.8 (7.0) | Matched | 66.9 (6.9) | 67.1 (6.9) | Matched |
Age at menarche, y | 12.5 (1.4) | 12.6 (1.4) | 0.07 | 12.6 (1.4) | 12.7 (1.4) | 0.27 |
Age at first birth among parous, y | 25.3 (3.4) | 25.0 (3.1) | 0.03 | 25.0 (3.2) | 24.8 (3.2) | 0.69 |
Age at menopause, y2 | 49.6 (4.3) | 49.1 (4.7) | 0.001 | 50.2 (4.1) | 49.3 (4.6) | 0.002 |
Parity among parous | 3.1 (1.5) | 3.2 (1.5) | 0.16 | 3.1 (1.4) | 3.1 (1.6) | 0.63 |
BMI at age 18, kg/m2 | 21.1 (2.7) | 21.3 (2.8) | 0.03 | 21.0 (2.8) | 21.2 (2.7) | 0.36 |
BMI at blood collection, kg/m2 | 25.5 (4.5) | 25.4 (4.6) | 0.33 | 26.9 (4.9) | 26.4 (4.8) | 0.18 |
Alcohol intake at blood collection, g/d3 | 5.5 (9.7) | 5.0 (8.8) | 0.09 | 7.3 (12.7) | 6.2 (10.5) | 0.19 |
% | % | |||||
Family history of breast cancer in 1st degree relatives, % | 15 | 10 | <0.0001 | 22 | 15 | 0.02 |
History of benign breast disease, % | 54 | 46 | <0.0001 | 64 | 61 | 0.39 |
Current use of multivitamins, %4 | 39 | 39 | 0.84 | 65 | 59 | 0.09 |
Postmenopausal women, % | 67 | 67 | Matched | 98 | 98 | Matched |
Recent use of postmenopausal hormones, %2 | 53 | 53 | Matched | 69 | 69 | Matched |
Plasma Concentrations | Mean (SD) | Mean (SD) | ||||
Folate (ng/mL) | 10.7 (8.9) | 10.8 (8.8) | 0.84 | 31.3 (17.1) | 29.7 (15.9) | 0.17 |
Vitamin B12 (pg/mL) | 489 (229) | 483 (217) | 0.39 | 624 (358) | 634 (351) | 0.71 |
Pyridoxal 5′-phosphate (pmol/mL) | 65.4 (78.5) | 66.7 (79.8) | 0.61 | 79.0 (92.3) | 75.8 (85.2) | 0.63 |
Homocysteine (nmol/mL) | 11.5 (3.7) | 11.3 (3.4) | 0.09 | 11.0 (3.1) | 10.8 (3.0) | 0.31 |
Cysteine (nmol/mL) | 275.5 (60.9) | 276.7 (61.8) | 0.23 | 239.2 (13.7) | 238.2 (13.3) | 0.22 |
Cysteinylglycine (nmol/mL) | 193.1 (55.1) | 193.0 (53.4) | 0.99 | 192.4 (53.1) | 193.0 (48.7) | 0.97 |
Abbreviations: SD = standard deviation; BMI = body mass index; Matched = values were similar because of matching.
p Values are from the paired t-test for continuous variable or from the Mantel–Haenszel test for categorical variables. All p values were two-sided.
Among postmenopausal women only.
From the 1990 FFQ for the 1990 blood collection or from the 1998 FFQ for the 2000 blood collection.
At the 1990 blood collection (from the 1990 FFQ) or at the 2000 blood collection (from the 2000 biennial questionnaire).
Plasma measures of folate, B12 and PLP were modestly correlated with each other (r = 0.3–0.5), homocysteine was weakly inversely correlated with folate, B12 and PLP, and cysteine was only correlated with homocysteine and cysteinylglycine (Supporting Information Table S1). On average, significant correlations between B-vitamin plasma measures and dietary intakes were 0.1 to 0.5, with the strongest correlations being plasma folate with total intake of folate and plasma PLP with total intake of B6.
The mean time between the two blood collections was 11.1 years (range = 9.4–13). Plasma measures had variable reproducibility over the approximately 11-year period. The highest ICC values were observed with vitamin B12 (0.51, 95% CI = 0.46–0.56) and cysteinylglycine (0.55, 95% CI = 0.50–0.60), with slightly lower values for PLP (0.47, 95% CI = 0.41–0.52), homocysteine (0.34, 95% CI = 0.28–0.41), and cysteine (0.44, 95% CI = 0.38–0.50). The ICC for folate was not calculated due to the three-fold increase in folate levels between the two blood collections. Spearman correlations over the 11-year interval were higher than ICC’s for all plasma measures: 0.35 for folate, 0.57 for B12, 0.49 for PLP, 0.36 for homocysteine, 0.51 for cysteine and 0.56 for cysteinylglycine.
No significant associations were seen for plasma folate, B12, PLP, homocysteine or cysteine with overall breast cancer risk (Table 2). The RR for the highest versus the lowest quintile ranged from 0.91 (95% CI = 0.73–1.13) for PLP to 1.17 (95% CI = 0.93–1.46) for homocysteine. None of the associations were significant when potential non-linearity was evaluated (not shown). Estimates were similar when we mutually adjusted for the other plasma measures and when we restricted to only those that had fasted ≥8 h prior to blood donation. No significant associations were seen when restricted to in situ or invasive breast cancer cases. None of the plasma B-vitamins or metabolites were significantly associated with breast cancer by ER/PR status. There were no significant associations for plasma folate, B12, PLP, cysteine and cysteinylglycine observed when assessed by molecular subtypes of breast cancer (Table 3). Homocysteine was not significantly associated when assessed by molecular subtypes when modeled in tertiles or in overall heterogeneity tests (P-heterogeneity >0.05). However, when modeled continuously per SD increase in natural log transformed concentration homocysteine was significantly associated with basal-like breast cancer (RR = 1.63, 95% CI = 1.23–2.17).
Table 2.
# Cases | Quintile of plasma measure1 | p for trend | p for heterogeneity | |||||
---|---|---|---|---|---|---|---|---|
1 (lowest) | 2 | 3 | 4 | 5 | ||||
Folate, ng/mL | <4.7 | 4.7 to 6.6 | 6.7 to 9.7 | 9.8 to 15.5 | ≥15.6 | |||
Overall | 1872 | 1.00 (ref.) | 0.94 (0.77–1.15) | 0.81 (0.65–1.00) | 0.85 (0.69–1.05) | 0.95 (0.77–1.17) | 0.81 | |
Insitu | 346 | 1.00 (ref.) | 0.72 (0.45–1.13) | 0.73 (0.44–1.22) | 1.00 (0.61–1.63) | 0.83 (0.51–1.33) | 0.96 | 0.37 |
Invasive | 1,502 | 1.00 (ref.) | 1.00 (0.79–1.27) | 0.82 (0.64–1.04) | 0.81 (0.64–1.03) | 0.97 (0.77–1.23) | 0.76 | |
ER+/PR+ | 913 | 1.00 (ref.) | 1.16 (0.85–1.57) | 0.81 (0.60–1.11) | 0.79 (0.57–1.08) | 1.01 (0.74–1.38) | 0.69 | 0.93 |
ER+/PR− | 199 | 1.00 (ref.) | 0.96 (0.48–1.90) | 0.76 (0.38–1.52) | 0.80 (0.40–1.59) | 1.08 (0.55–2.14) | 0.68 | |
ER−/PR− | 230 | 1.00 (ref.) | 1.03 (0.55–1.92) | 1.16 (0.60–2.22) | 1.08 (0.57–2.05) | 0.97 (0.52–1.81) | 0.90 | |
Vitamin B12, pg/mL | <311 | 311 to 403 | 404 to 497 | 498 to 618 | ≥619 | |||
Overall | 1862 | 1.00 (ref.) | 1.09 (0.88–1.35) | 1.11 (0.91–1.37) | 0.92 (0.75–1.15) | 1.06 (0.85–1.31) | 0.88 | |
Insitu | 344 | 1.00 (ref.) | 1.00 (0.61–1.64) | 0.85 (0.52–1.39) | 0.90 (0.54–1.51) | 0.85 (0.51–1.42) | 0.49 | 0.54 |
Invasive | 1,494 | 1.00 (ref.) | 1.08 (0.85–1.38) | 1.22 (0.96–1.54) | 0.93 (0.73–1.18) | 1.11 (0.87–1.41) | 0.81 | |
ER+/PR+ | 909 | 1.00 (ref.) | 1.15 (0.84–1.56) | 1.42 (1.05–1.91) | 1.06 (0.78–1.45) | 1.14 (0.83–1.56) | 0.74 | 0.34 |
ER+/PR− | 199 | 1.00 (ref.) | 0.72 (0.34–1.52) | 1.01 (0.50–2.03) | 0.94 (0.45–1.96) | 0.83 (0.40–1.71) | 0.90 | |
ER−/PR− | 227 | 1.00 (ref.) | 0.96 (0.49–1.87) | 0.57 (0.30–1.11) | 0.60 (0.33–1.11) | 0.95 (0.51–1.76) | 0.63 | |
Pyridoxal 5′-phosphate, pmol/mL | <24.4 | 24.4 to 35.0 | 35.1 to 50.0 | 50.1 to 82.6 | ≥82.7 | |||
Overall | 1872 | 1.00 (ref.) | 0.91 (0.74–1.11) | 0.93 (0.75–1.15) | 0.81 (0.66–1.01) | 0.91 (0.73–1.13) | 0.56 | |
Insitu | 347 | 1.00 (ref.) | 0.86 (0.52–1.41) | 0.96 (0.58–1.59) | 0.82 (0.47–1.41) | 0.93 (0.55–1.57) | 0.90 | 0.99 |
Invasive | 1,501 | 1.00 (ref.) | 0.90 (0.71–1.13) | 0.89 (0.70–1.14) | 0.81 (0.63–1.03) | 0.86 (0.67–1.11) | 0.37 | |
ER+/PR+ | 912 | 1.00 (ref.) | 0.89 (0.66–1.21) | 0.91 (0.66–1.25) | 0.85 (0.62–1.18) | 0.87 (0.62–1.21) | 0.56 | 0.96 |
ER+/PR− | 200 | 1.00 (ref.) | 1.02 (0.49–2.12) | 1.20 (0.60–2.42) | 0.77 (0.38–1.58) | 0.72 (0.35–1.46) | 0.18 | |
ER−/PR− | 229 | 1.00 (ref.) | 0.89 (0.51–1.55) | 0.92 (0.49–1.74) | 0.87 (0.47–1.59) | 0.86 (0.44–1.69) | 0.75 | |
Homocysteine, nmol/mL | <8.6 | 8.6 to 9.8 | 9.9 to 11.3 | 11.4 to 13.3 | ≥13.4 | |||
Overall | 1863 | 1.00 (ref.) | 1.11 (0.89–1.38) | 0.98 (0.79–1.22) | 1.12 (0.90–1.40) | 1.17 (0.93–1.46) | 0.20 | |
Insitu | 345 | 1.00 (ref.) | 0.96 (0.57–1.61) | 0.89 (0.55–1.44) | 0.96 (0.58–1.57) | 0.92 (0.54–1.58) | 0.79 | 0.97 |
Invasive | 1,495 | 1.00 (ref.) | 1.13 (0.89–1.45) | 0.98 (0.76–1.26) | 1.12 (0.87–1.45) | 1.19 (0.92–1.54) | 0.22 | |
ER+/PR+ | 909 | 1.00 (ref.) | 0.92 (0.68–1.25) | 0.99 (0.72–1.35) | 1.05 (0.76–1.45) | 1.23 (0.88–1.71) | 0.10 | 0.45 |
ER+/PR− | 198 | 1.00 (ref.) | 1.46 (0.73–2.92) | 0.78 (0.38–1.60) | 1.25 (0.58–2.70) | 1.37 (0.63–2.98) | 0.56 | |
ER−/PR− | 230 | 1.00 (ref.) | 1.61 (0.78–3.35) | 1.54 (0.73–3.27) | 1.24 (0.59–2.61) | 1.24 (0.60–2.58) | 0.81 | |
Cysteine, nmol/mL2 | ||||||||
Overall | 1872 | 1.00 (ref.) | 1.18 (0.95–1.47) | 1.12 (0.90–1.41) | 1.01 (0.79–1.30) | 0.97 (0.73–1.30) | 0.93 | |
Insitu | 347 | 1.00 (ref.) | 1.02 (0.61–1.69) | 1.18 (0.69–2.00) | 0.95 (0.52–1.75) | 1.14 (0.55–2.37) | 0.54 | 0.78 |
Invasive | 1,501 | 1.00 (ref.) | 1.21 (0.95–1.55) | 1.09 (0.85–1.41) | 0.97 (0.74–1.28) | 0.91 (0.66–1.26) | 0.65 | |
ER+/PR+ | 912 | 1.00 (ref.) | 1.42 (1.04–1.94) | 1.27 (0.92–1.76) | 1.00 (0.69–1.44) | 1.03 (0.68–1.56) | 0.83 | 0.03 |
ER+/PR− | 200 | 1.00 (ref.) | 1.67 (0.80–3.49) | 0.95 (0.46–1.98) | 1.51 (0.68–3.36) | 0.80 (0.30–2.13) | 0.44 | |
ER−/PR− | 229 | 1.00 (ref.) | 0.36 (0.18–0.75) | 0.57 (0.27–1.22) | 0.65 (0.33–1.29) | 0.62 (0.25–1.56) | 0.41 | |
Cysteinylglycine, nmol/mL | <174.8 | 174.8 to 198.9 | 199.0 to 232.6 | 232.7 to 515.8 | ≥515.9 | |||
Overall | 1846 | 1.00 (ref.) | 0.95 (0.77–1.18) | 0.80 (0.64–1.01) | 0.93 (0.75–1.17) | 0.98 (0.78–1.23) | 0.96 | |
Insitu | 344 | 1.00 (ref.) | 1.03 (0.62–1.72) | 0.80 (0.46–1.40) | 1.26 (0.71–2.25) | 0.79 (0.45–1.41) | 0.57 | 0.24 |
Invasive | 1,478 | 1.00 (ref.) | 0.93 (0.73–1.18) | 0.82 (0.63–1.05) | 0.86 (0.67–1.11) | 1.03 (0.79–1.33) | 0.81 | |
ER+/PR+ | 902 | 1.00 (ref.) | 0.99 (0.72–1.36) | 0.78 (0.56–1.08) | 0.87 (0.63–1.21) | 0.98 (0.70–1.37) | >0.89 | 0.89 |
ER+/PR− | 198 | 1.00 (ref.) | 0.73 (0.38–1.39) | 0.86 (0.44–1.71) | 0.94 (0.47–1.90) | 1.05 (0.53–2.08) | 0.69 | |
ER−/PR− | 227 | 1.00 (ref.) | 0.65 (0.33–1.27) | 0.91 (0.44–1.89) | 0.75 (0.37–1.51) | 1.14 (0.54–2.43) | 0.42 |
Adjustments for age at menarche (continuous variable), parity/age at first birth (nulliparous, 1–2 children/first birth <25 years, 1–2 children/first birth 25+ years, ≥3 children/first birth <25 years, ≥3 children/first birth 25+ years), age at menopause (<50, ≥50 to <55, or ≥ 55 years), family history of breast cancer in mother or a sister (yes or no), history of benign breast disease (yes or no), height (continuous), body mass index at 18 (<21, 21 to <23, 23+), weight change from age 18 (continuous) and alcohol intake at blood collection (continuous) in 1990.
Quintile cut points: <246.9, 246.9 to 273.4, 273.5 to 300.2, 300.3 to 345.0, ≥345.1 Batch 1–3; <222.8, 222.8 to 230.8, 230.9 to 238.3, 238.4 to 245.4, ≥245.5 Batch 4.
Table 3.
# Cases | Tertile of plasma measure1 | P for trend | P for heterogeneity | Per 1-SD increase in natural log-transformed concentrations1 | |||
---|---|---|---|---|---|---|---|
1 (lowest) | 2 | 3 | |||||
Folate, ng/mL | <6.0 | 6.0 to 11.2 | ≥11.3 | ||||
Luminal A | 510 | 1.00 (ref.) | 0.87 (0.68–1.12) | 1.05 (0.82–1.34) | 0.47 | 0.41 | 1.01 (0.91–1.12) |
Luminal B | 182 | 1.00 (ref.) | 0.71 (0.48–1.03) | 0.79 (0.54–1.16) | 0.37 | 0.93 (0.79–1.10) | |
HER 2 Type | 50 | 1.00 (ref.) | 0.82 (0.40–1.68) | 0.95 (0.47–1.93) | 0.99 | 0.89 (0.66–1.20) | |
Basal Like | 42 | 1.00 (ref.) | 1.47 (0.68–3.17) | 1.14 (0.49–2.63) | 0.94 | 0.94 (0.68–1.30) | |
Vitamin B12, pg/mL | <375 | 375 to 530 | ≥531 | ||||
Luminal A | 508 | 1.00 (ref.) | 1.11 (0.87–1.42) | 1.04 (0.81–1.33) | 0.83 | 0.81 | 1.04 (0.94–1.16) |
Luminal B | 182 | 1.00 (ref.) | 0.94 (0.64–1.36) | 0.93 (0.64–1.36) | 0.72 | 1.05 (0.90–1.22) | |
HER 2 Type | 50 | 1.00 (ref.) | 0.83 (0.40–1.75) | 1.20 (0.61–2.38) | 0.52 | 1.12 (0.84–1.49) | |
Basal Like | 42 | 1.00 (ref.) | 0.80 (0.37–1.70) | 0.72 (0.33–1.54) | 0.40 | 0.86 (0.63–1.17) | |
Pyridoxal 5′-phosphate, pmol/mL | <31.1 | 31.1 to 57.7 | ≥57.8 | ||||
Luminal A | 510 | 1.00 (ref.) | 0.86 (0.68–1.10) | 0.88 (0.68–1.12) | 0.40 | 0.45 | 0.98 (0.88–1.08) |
Luminal B | 182 | 1.00 (ref.) | 0.90 (0.62–1.30) | 0.82 (0.56–1.21) | 0.34 | 0.92 (0.79–1.08) | |
HER 2 Type | 50 | 1.00 (ref.) | 0.60 (0.30–1.20) | 0.57 (0.28–1.16) | 0.18 | 0.76 (0.55–1.05) | |
Basal Like | 41 | 1.00 (ref.) | 1.72 (0.77–3.87) | 1.66 (0.73–3.78) | 0.34 | 1.02 (0.74–1.40) | |
Homocysteine, nmol/mL | <9.5 | 9.5 to 11.8 | ≥11.9 | ||||
Luminal A | 509 | 1.00 (ref.) | 0.96 (0.75–1.22) | 1.02 (0.80–1.31) | 0.81 | 0.29 | 0.96 (0.86–1.06) |
Luminal B | 182 | 1.00 (ref.) | 1.08 (0.73–1.60) | 1.22 (0.83–1.79) | 0.31 | 1.11 (0.95–1.30) | |
HER 2 Type | 50 | 1.00 (ref.) | 0.51 (0.25–1.05) | 0.60 (0.30–1.20) | 0.16 | 0.85 (0.63–1.15) | |
Basal Like | 42 | 1.00 (ref.) | 0.85 (0.36–2.04) | 1.56 (0.73–3.33) | 0.18 | 1.63 (1.23–2.17) | |
Cysteine, nmol/mL2 | |||||||
Luminal A | 510 | 1.00 (ref.) | 1.00 (0.79–1.28) | 0.86 (0.66–1.11) | 0.21 | 0.57 | 0.99 (0.89–1.10) |
Luminal B | 182 | 1.00 (ref.) | 1.23 (0.85–1.80) | 0.88 (0.58–1.35) | 0.43 | 1.09 (0.93–1.29) | |
HER 2 Type | 50 | 1.00 (ref.) | 0.84 (0.42–1.68) | 0.66 (0.30–1.41) | 0.28 | 0.86 (0.63–1.18) | |
Basal Like | 42 | 1.00 (ref.) | 1.57 (0.67–3.69) | 1.84 (0.77–4.38) | 0.20 | 1.24 (0.89–1.72) | |
Cysteinylglycine, nmol/mL2 | <167.3 | 167.3 to 208.7 | ≥208.8 | ||||
Luminal A | 505 | 1.00 (ref.) | 0.78 (0.61–1.00) | 0.87 (0.68–1.11) | 0.27 | 0.85 | 0.97 (0.88–1.08) |
Luminal B | 181 | 1.00 (ref.) | 0.84 (0.57–1.24) | 1.06 (0.73–1.54) | 0.71 | 1.02 (0.87–1.19) | |
HER 2 Type | 50 | 1.00 (ref.) | 1.22 (0.60–2.47) | 1.13 (0.55–2.34) | 0.76 | 1.04 (0.77–1.41) | |
Basal Like | 42 | 1.00 (ref.) | 0.91 (0.41–2.04) | 1.13 (0.53–2.42) | 0.73 | 1.17 (0.84–1.62) |
Adjustments for matching factors, age at menarche (continuous variable), parity/age at first birth (nulliparous, 1–2 children/first birth <25 years, 1–2 children/first birth 25+ years, ≥3 children/first birth <25 years, ≥3 children/first birth 25+ years), age at menopause (<50, ≥50 to <55, or ≥ 55 years), family history of breast cancer in mother or a sister (yes or no), history of benign breast disease (yes or no), height (continuous), body mass index at 18 (<21, 21 to <23, 23+), weight change from age 18 (continuous) and alcohol intake at blood collection (continuous) in 1990.
Tertile cut points: <264.4, 264.4 to 313.0, ≥313.1 Batch 1–3; <228.9, 228.9 to 240.9, ≥241.0 Batch 4.
There were no significant associations for plasma folate, B12, homocysteine, cysteine, or cysteinylglycine at 1990 blood collection with cases diagnosed within 10 years of blood collection (proximate), 1990 blood collection with cases diagnosed >10 years of blood collection (distant), or the 2000 blood sample (proximate cases, blood sample post fortification) with invasive breast cancer (Table 4). For plasma PLP, there was a significant inverse association observed for the proximate 1990 blood sample and invasive breast cancer (RR top vs. bottom tertile = 0.79, 95% CI = 0.64–0.97, p-trend = 0.04). However, this inverse association was not seen after fortification (2000 blood collection) or with distant invasive breast cancer cases (1990 blood collection).
Table 4.
# Cases | Tertile of plasma measure1 | p for trend | Per 1-SD increase in natural log-transformed concentrations1 | |||
---|---|---|---|---|---|---|
1 (lowest) | 2 | 3 | ||||
Folate, ng/mL | ||||||
<6.0 | 6.0 to 11.2 | ≥11.3 | ||||
1990 blood: 1990–2000 follow-up | 1,041 | 1.00 (ref.) | 0.95 (0.77–1.16) | 0.93 (0.75–1.16) | 0.56 | 0.92 (0.83–1.01) |
2000–2006 follow-up | 450 | 1.00 (ref.) | 0.79 (0.57–1.10) | 0.97 (0.72–1.31) | 0.92 | 1.03 (0.92–1.15) |
<21.9 | 21.9 to 31.9 | ≥32.0 | ||||
2000 blood: 2000–2006 follow-up | 282 | 1.00 (ref.) | 1.14 (0.76–1.71) | 1.17 (0.79–1.74) | 0.47 | 1.14 (0.97–1.34) |
Vitamin B12, pg/mL | ||||||
<375 | 375 to 530 | ≥531 | ||||
1990 blood: 1990–2000 follow-up | 1,039 | 1.00 (ref.) | 0.93 (0.75–1.14) | 1.01 (0.81–1.24) | 0.87 | 1.05 (0.96–1.16) |
2000–2006 follow-up | 449 | 1.00 (ref.) | 1.24 (0.90–1.71) | 1.01 (0.75–1.35) | 0.98 | 1.03 (0.92–1.15) |
<425 | 425 to 736 | ≥737 | ||||
2000 blood: 2000–2006 follow-up | 282 | 1.00 (ref.) | 1.26 (0.86–1.85) | 0.87 (0.58–1.32) | 0.40 | 1.02 (0.87–1.20) |
Pyridoxal 5′-phosphate, pmol/mL | ||||||
<31.1 | 31.1 to 57.7 | ≥57.8 | ||||
1990 blood: 1990–2000 follow-up | 1,041 | 1.00 (ref.) | 0.86 (0.70–1.06) | 0.79 (0.64–0.97) | 0.04 | 0.92 (0.85–1.00) |
2000–2006 follow-up | 449 | 1.00 (ref.) | 0.98 (0.72–1.32) | 1.15 (0.85–1.57) | 0.33 | 1.08 (0.95–1.22) |
2000 blood: 2000–2006 follow-up | 282 | 1.00 (ref.) | 0.85 (0.56–1.30) | 1.08 (0.72–1.61) | 0.46 | 1.04 (0.88–1.23) |
Homocysteine, nmol/mL | ||||||
<9.5 | 9.5 to 11.8 | ≥11.9 | ||||
1990 blood: 1990–2000 follow-up | 1,037 | 1.00 (ref.) | 0.90 (0.73–1.12) | 1.03 (0.84–1.26) | 0.67 | 1.03 (0.95–1.12) |
2000–2006 follow-up | 449 | 1.00 (ref.) | 0.88 (0.65–1.20) | 1.21 (0.88–1.67) | 0.20 | 1.09 (0.95–1.26) |
2000 blood: 2000–2006 follow-up | 281 | 1.00 (ref.) | 1.08 (0.74–1.59) | 1.12 (0.74–1.69) | 0.59 | 1.00 (0.84–1.19) |
Cysteine, nmol/mL | ||||||
<264.4 | 264.4 to 313.0 | ≥313.1 | ||||
1990 blood: 1990–2000 follow-up | 1,042 | 1.00 (ref.) | 0.99 (0.80–1.22) | 0.85 (0.68–1.06) | 0.12 | 0.95 (0.86–1.04) |
<228.9 | 228.9–240.9 | ≥241.0 | ||||
2000–2006 follow-up | 450 | 1.00 (ref.) | 1.12 (0.83–1.51) | 0.95 (0.68–1.34) | 0.73 | 1.08 (0.94–1.23) |
2000 blood: 2000–2006 follow-up | 282 | 1.00 (ref.) | 1.30 (0.83–2.02) | 1.09 (0.70–1.69) | 0.85 | 1.13 (0.95–1.35) |
Cysteinylglycine, nmol/mL | ||||||
<167.3 | 167.3 to 208.7 | ≥208.8 | ||||
1990 blood: 1990–2000 follow-up | 1,018 | 1.00 (ref.) | 0.91 (0.74–1.11) | 1.00 (0.81–1.23) | 0.98 | 1.00 (0.91–1.09) |
2000–2006 follow-up | 450 | 1.00 (ref.) | 0.75 (0.55–1.04) | 1.00 (0.74–1.35) | 0.92 | 1.02 (0.90–1.15) |
2000 blood: 2000–2006 follow-up | 281 | 1.00 (ref.) | 0.74 (0.50–1.09) | 0.88 (0.59–1.32) | 0.57 | 0.97 (0.83–1.15) |
Adjustments for matching factors, age at menarche (continuous variable), parity/age at first birth (nulliparous, 1–2 children/first birth <25 years, 1–2 children/first birth 25+ years, ≥3 children/first birth <25 years, ≥3 children/first birth 25+ years), age at menopause (<50, ≥50 to <55, or ≥ 55 years), family history of breast cancer in mother or a sister (yes or no), history of benign breast disease (yes or no), height (continuous), body mass index at 18 (<21, 21 to <23, 23+), weight change from age 18 (continuous) and alcohol intake at blood collection (continuous) in 1990, or in 2000 dependent on models.
No significant associations were observed for the average of the 1990 and 2000 plasma measures with breast cancer risk; though a borderline positive association was noted for the continuous folate average and breast cancer (Table 5). In updated plasma measure analyses, no association was seen for plasma folate or other plasma measures, with the exception of continuous (per1 SD increase in natural log transformed plasma measure) plasma homocysteine, where an 8% increase in breast cancer risk was seen.
Table 5.
Tertile of plasma measure | p for trend | Per 1-SD increase in natural log-transformed concentrations | |||
---|---|---|---|---|---|
1 (lowest) | 2 | 3 | |||
Average of 1990 and 2000 plasma measures1 | |||||
Folate, ng/mL | <11.6 | 11.6 to 18.4 | ≥18.5 | ||
1.00 (ref) | 0.79 (0.53, 1.17) | 1.38 (0.94, 2.03) | 0.03 | 1.20 (1.00, 1.44) | |
Vitamin B12, pg/mL | <415 | 415 to 612 | ≥613 | ||
1.00 (ref) | 1.02 (0.70, 1.49) | 0.97 (0.66, 1.41) | 0.83 | 1.03 (0.88, 1.22) | |
Pyridoxal 5′-phosphate, pg/mL | <33.7 | 33.7 to 57.7 | ≥57.8 | ||
1.00 (ref) | 0.79 (0.54, 1.15) | 1.06 (0.72, 1.54) | 0.59 | 1.06 (0.88, 1.27) | |
Homocysteine, nmol/mL | <9.5 | 9.5 to 11.2 | ≥11.3 | ||
1.00 (ref) | 0.80 (0.54, 1.19) | 1.17 (0.80, 1.73) | 0.34 | 1.09 (0.88, 1.33) | |
Cysteine, nmol/mL | <229.6 | 229.6 to 240.6 | ≥240.7 | ||
1.00 (ref) | 1.36 (0.92, 2.00) | 1.15 (0.75, 1.77) | 0.53 | 1.19 (0.97, 1.46) | |
Cysteinylglycine, nmol/mL | <171.0 | 171.0 to 210.3 | ≥210.4 | ||
1.00 (ref) | 0.78 (0.54, 1.15) | 0.84 (0.57, 1.22) | 0.37 | 0.93 (0.78, 1.11) | |
Updated plasma measures2 | |||||
Folate, ng/mL | 1.00 (ref) | 1.04 (0.88, 1.22) | 0.93 (0.79, 1.10) | 0.52 | 0.97 (0.90, 1.04) |
Vitamin B12, pg/mL | 1.00 (ref) | 1.11 (0.94, 1.30 | 0.95 (0.80, 1.12) | 0.49 | 1.02 (0.94, 1.09) |
Pyridoxal 5′-phosphate, pg/mL | 1.00 (ref) | 0.95 (0.81, 1.12) | 0.93 (0.79, 1.10) | 0.42 | 0.98 (0.92, 1.05) |
Homocysteine, nmol/mL | 1.00 (ref) | 0.91 (0.77, 1.08) | 1.16 (0.98, 1.37) | 0.08 | 1.08 (1.01, 1.15) |
Cysteine, nmol/mL | 1.00 (ref) | 1.07 (0.91, 1.27) | 0.99 (0.83, 1.18) | 0.15 | 0.98 (0.92, 1.06) |
Cysteinylglycine, nmol/mL | 1.00 (ref) | 0.84 (0.71, 0.98) | 0.89 (0.76, 1.06) | 0.37 | 0.95 (0.88, 1.02) |
Unconditional logistic regression using the average of the 1990 and 2000 plasma measures among only those with two blood collections (2000–2006 follow-up; Cases/Controls:367/367) with adjustments for matching factors, age at menarche (continuous variable), parity/age at first birth (nulliparous, 1–2 children/first birth <25 years, 1–2 children/first birth 25+ years, ≥3 children/first birth <25 years, ≥3 children/first birth 25+ years), age at menopause (<50, ≥50 to <55, or ≥ 55 years), family history of breast cancer in mother or a sister (yes or no), history of benign breast disease (yes or no), height (continuous), body mass index at 18 (<21, 21 to <23, 23+), weight change from age 18 (continuous) and alcohol intake at blood collection (continuous) in both 1990 and 2000.
Repeated measures logistic regression, where 1990 plasma measure used for 1990–2000 time period and the 2000 plasma measure for the 2000–2006 time period (Cases/Controls:1626/1855). Controls from the 2000–2006 case–control pairs were allowed to contribute to both the 1990–2000 and 2000–2006 time periods. Models were adjusted for matching factors, age at menarche (continuous variable), parity/age at first birth (nulliparous, 1–2 children/first birth <25 years, 1–2 children/first birth 25+ years, ≥3 children/first birth <25 years, ≥3 children/first birth 25+ years), age at menopause (<50, ≥50 to <55, or ≥ 55 years), family history of breast cancer in mother or a sister (yes or no), history of benign breast disease (yes or no), height (continuous), body mass index at 18 (<21, 21 to <23, 23+), weight change from age 18 (continuous) and alcohol intake (continuous), updated at time of blood collection when relevant. See Table 4 for blood collection specific tertile cut points.
Associations of folate and other plasma measures (per SD of natural log–transformed plasma measure) did not vary by age at blood donation (Table 6). For plasma folate, B12 and homocysteine, associations varied by smoking status; though no strata reached statistical significance. The B12, PLP and cysteine associations varied significantly by BMI, although only B12 reached significance in stratified analyses. For example, for B12, among those with BMI <25 the relative risk was 0.97 (95% CI = 0.88–1.06) and among those with BMI ≥25 the relative risk was 1.17 (95% CI = 1.05–1.30, p-interaction = <0.0001). For all plasma measures, associations did not vary by alcohol use (p-interaction >0.05 for all). Lastly, associations did not significantly vary when restricted to nonmultivitamin users (data not shown).
Table 6.
Plasma measure | Cases/ Controls | Per 1-SD increase in natural log-transformed concentrations RR (95% CI)1 | Cases/Controls | Per 1-SD increase in natural log-transformed concentrations RR (95% CI)1 | p for interaction |
---|---|---|---|---|---|
Age at 1990 blood <57 years | Age at 1990 blood ≥57 years | ||||
Folate, ng/mL | 704/935 | 0.98 (0.88,1.08) | 796/935 | 0.92 (0.83,1.01) | 0.10 |
Vitamin B12, pg/mL | 703/933 | 0.99 (0.89,1.09) | 793/932 | 1.10 (1.00,1.21) | 0.07 |
Pyridoxal 5′-phosphate, pmol/mL | 705/938 | 0.99 (0.89,1.10) | 796/935 | 0.93 (0.84,1.02) | 0.06 |
Homocysteine, nmol/mL | 703/930 | 1.07 (0.96,1.19) | 787/932 | 1.03 (0.94,1.14) | 0.70 |
Cysteine, nmol/mL | 705/936 | 0.99 (0.89,1.11) | 796/935 | 0.99 (0.89,1.10) | 0.62 |
Cysteinylglycine, nmol/mL | 693/920 | 0.98 (0.89,1.09) | 773/919 | 1.02 (0.93,1.13) | 0.28 |
Never/Past Smoker at 1990 Blood | Current Smoker at 1990 blood | ||||
Folate, ng/mL | 1300/1657 | 0.99 (0.91,1.07) | 199/207 | 0.81 (0.66,1.00) | 0.04 |
Vitamin B12, pg/mL | 1296/1652 | 1.08 (1.00,1.16) | 199/207 | 0.84 (0.69,1.04) | 0.003 |
Pyridoxal 5’-phosphate, pmol/mL | 1301/1660 | 0.98 (0.91,1.06) | 199/207 | 0.93 (0.75,1.14) | 0.59 |
Homocysteine, nmol/mL | 1292/1650 | 1.01 (0.94,1.10) | 197/206 | 1.17 (0.97,1.43) | 0.03 |
Cysteine, nmol/mL | 1301/1658 | 1.00 (0.92,1.08) | 199/207 | 0.93 (0.74,1.18) | 0.76 |
Cysteinylglycine, nmol/mL | 1272/1629 | 0.99 (0.91,1.06) | 193/204 | 1.09 (0.88,1.33) | 0.17 |
BMI <25 kg/m2 | BMI ≥25 kg/m2 | ||||
Folate, ng/mL | 789/1046 | 0.94 (0.85,1.03) | 711/824 | 0.97 (0.87,1.09) | 0.18 |
Vitamin B12, pg/mL | 786/1042 | 0.97 (0.88,1.06) | 710/823 | 1.17 (1.05,1.30) | <0.0001 |
Pyridoxal 5’-phosphate, pmol/mL | 790/1046 | 0.91 (0.82,1.00) | 711/827 | 1.05 (0.94,1.17) | 0.002 |
Homocysteine, nmol/mL | 781/1042 | 1.04 (0.94,1.14) | 709/820 | 1.07 (0.96,1.19) | 0.56 |
Cysteine, nmol/mL | 790/1046 | 0.92 (0.84,1.02) | 711/825 | 1.07 (0.95,1.19) | 0.0009 |
Cysteinylglycine, nmol/mL | 771/1028 | 1.04 (0.95,1.15) | 695/811 | 0.96 (0.87,1.07) | 0.13 |
Alcohol <10 g/day | Alcohol ≥10 g/day | ||||
Folate, ng/mL | 1212/1535 | 0.96 (0.88,1.03) | 288/335 | 0.97 (0.81,1.15) | 0.81 |
Vitamin B12, pg/mL | 1208/1531 | 1.03 (0.95,1.11) | 288/334 | 1.12 (0.95,1.33) | 0.16 |
Pyridoxal 5’-phosphate, pmol/mL | 1212/1538 | 0.97 (0.90,1.05) | 289/335 | 0.95 (0.80,1.13) | 0.76 |
Homocysteine, nmol/mL | 1206/1529 | 1.04 (0.96,1.13) | 284/333 | 1.10 (0.93,1.30) | 0.30 |
Cysteine, nmol/mL | 1212/1536 | 0.98 (0.91,1.07) | 289/335 | 1.05 (0.87,1.27) | 0.29 |
Cysteinylglycine, nmol/mL | 1188/1509 | 1.00 (0.93,1.08) | 278/330 | 0.99 (0.83,1.18) | 0.99 |
Adjustments for matching factors, age at menarche (continuous variable), parity/age at first birth (nulliparous, 1–2 children/first birth <25 years, 1–2 children/first birth 25+ years, ≥3 children/first birth <25 years, ≥3 children/first birth 25+ years), age at menopause (<50, ≥50 to <55, or ≥ 55 years), family history of breast cancer in mother or a sister (yes or no), history of benign breast disease (yes or no), height (continuous), body mass index at 18 (<21, 21 to <23, 23+), weight change from age 18 (continuous) and alcohol intake at blood collection (continuous) in 1990.
Discussion
Overall, we found that folate, B12, PLP, homocysteine, or cysteine were not associated with breast cancer risk. Additionally, there were no consistent associations when exposure was assessed either before or after fortification, with proximal, distal, or the long term average exposures, or when cases were defined by specific tumor molecular subtypes. There was some suggestion that higher plasma levels of folate and B12, which in turn would lower homocysteine levels, may be beneficial among smokers.
Excluding the prior two studies conducted in the Nurses’ Health Study with follow-up to 1996, our findings are in line with the majority of other prospective studies finding no association of plasma B-vitamins and risk of breast cancer overall. Of seven studies that examined plasma folate, six found no association,18,19,35–38 and one found a positive overall association.39 Kim (2016) was a high risk population of BRCA1/2 carriers which could potentially explain the difference.39 Five studies examined plasma B12, all of which found no overall association.18,19,35,37,39 PLP was evaluated in four prior studies, of which three found no association,18,35,39 and one found an inverse overall association.37 Two examined homocysteine and found no association.35,40 One examined cysteine, finding a positive association with breast cancer.40 Additionally, to date, gene variants associated with vitamin B-related nutrient levels41,42 have not been associated with breast cancer in large GWAS studies.43
The two studies published previously in the NHS have cases that overlap with this analysis.44,45 Briefly, we previously observed inverse associations for cysteine,45 borderline significant inverse associations for folate, B12 and PLP, and no association for homocysteine with breast cancer.44 When stratified by menopausal status, inverse associations with PLP were stronger in postmenopausal women, whereas B12 was inversely associated with risk among premenopausal women.44 When stratified by alcohol intake, women who consumed at least 15 g alcohol per day had significant inverse associations with plasma folate (RR top vs. bottom = 0.11, 95% CI = 0.02–0.59).44 This current analysis expands on the follow-up period with an additional 10 years of follow-up (1990–2006) and an additional 1,162 cases, and we found no association overall for any of the five measures. We did observe inverse associations for PLP for our proximal 1990 blood sample analysis consistent with the prior analysis.
Prior literature on interactions is limited and inconsistent. In several studies, associations did not vary by alcohol intake18,40 similar to our results; conversely, one found B12 associations were positive among non-drinkers37 and one noting inverse for PLP among those consuming > 7 g/day.37 We observed that among current smokers, higher plasma folate was inversely associated and conversely, homocysteine was suggestive of a positive association with breast cancer risk. B12 had a significant interaction term, but the inverse association in stratified analysis among current smokers was not significant. In the two relatively small studies that assessed smoking,18,40 no differences in associations were observed by smoking status. Smokers have been shown to have lower systemic levels of folate,46–49 B1246,47 and B6,47,48 and higher homocysteine50 levels than non-smokers. The lower folate and B6 status persists after accounting for diet47–49 suggesting that smoking alters the metabolism of folate. Without B12, folate remains in the 5-methyl THF form unable to donate methyl groups or return to the folate cycle.2 One study found that smokers had different proportions of folate forms,47 with the majority of folate as 5-methyl THF, consistent with the “methyl trap” mechanism. Therefore, it is possible that among smokers having higher folate levels may partially mitigate this compared to the non-smokers.
The main strengths of the study are our ability to examine plasma B-vitamin levels before and after fortification due to the two blood collections and the ability to examine molecular subtypes with the unique tumor tissue resources. Other strengths of the study include its prospective design with overall large sample size, long-term follow-up, high follow-up rates, and accounting for known breast cancer risk factors.
Several of the stratified analyses were limited by small case numbers—particularly the low number of current smokers, and the small number of HER2 and basal breast cancer subtypes. There is also the potential for misclassification as a single blood sample was used to characterize a woman’s long-term levels. However, reported ICCs for women over 1–5 years ranged from 0.50 to 0.65,51,52 suggesting only moderate misclassification. Additionally, we had two blood samples about 10 years apart for a subset of the women. Lastly, folate was measured using two different methods – cases up until 2000 measured via radioassay and cases after 2000 measured via microbial assay. Microbial assays have a tendency to recover more folate than the radioimmunoassay and are considered the gold standard now; however, the two assays are highly correlated (R2 = 0.91).53 As case–control pairs after 2000 had both blood samples measured together, the difference between the 1990 and 2000 blood samples would not be due to the difference in assay methods. Additionally, the average batch recalibration should “adjust” for the difference in assay methods.
As our follow-up after the implementation of folic acid fortification was relatively short, (8 years after fortification implementation) we cannot exclude the possibility of an association between post-fortification plasma folate and breast cancers diagnosed 10–15+ years after fortification was implemented. Further, the number of cases diagnosed after fortification with a post-fortification blood sample may have limited our power to detect an association. Lastly, the metabolites measured were only from the folate cycle and the transulfuration pathway. Metabolites in the methionine cycle (i.e., SAM, S-adenosylhomocysteine [SAH]), which are directly involved in methylation and reflect the methylation potential (i.e., SAM/SAH)54 hypothesized to be the link between one-carbon metabolism and carcinogenesis, were not measured.
In summary, folate and other B-vitamins were not associated with breast cancer risk overall. There was some suggestion that higher plasma folate may be beneficial particularly to smokers. However, long term effects of folate fortification cannot be excluded, and additional large prospective studies are needed to confirm subgroup findings.
Supplementary Material
What’s new?
With concerns of increased cancer risk, there has been discussion as to whether countries should implement folic acid fortification programs. However, few prospective studies have examined plasma folate and other B-vitamins levels and breast cancer risk among populations whose food is fortified with folic acid. This study examines the association of plasma folate and other B-vitamins on breast cancer risk, prior to and after mandatory folic acid fortification of grain products in the U.S. Plasma folate, B12, PLP, homocysteine, cysteine, and cysteinylglycine were not significantly associated with breast cancer overall, before and after fortification, or with specific tumor molecular subtypes.
Acknowledgements
The authors thank the participants and staff of the NHS for their valuable contributions as well as the after state cancer registries for their help: AL, AZ, AR, CA, CO, CT, DE, FL, GA, ID, IL, IN IA, KY, LA, ME, MD, MI, NE, NH, NJ, NY, NC, ND, OH, OK, OR, PA, RI, SC, TN, TX, VA, WA and WY. The authors assume full responsibility for analyses and interpretation of these data.
Grant sponsor: Division of Cancer Prevention, National Cancer Institute; Grant numbers: P01 CA87969R01 CA124857R01 CA49449UM1 CA186107
Abbreviations:
- BMI
body mass index
- CI
confidence intervals
- CK5/6
cytokeratin 5/6
- EGFR
epidermal growth factor
- ER
estrogen receptor
- FFQ
food frequency questionnaires
- HER2
human epidermal growth factor 2
- ICC
intraclass correlation coefficients
- NHS
Nurses’ Health Study
- PR
progesterone receptor
- RR
relative risks
- SAM
S-adenosyl methionine
- SD
standard deviation
- THF
tetrahydrofolate
Footnotes
Additional Supporting Information may be found in the online version of this article.
Conflict of interest: The authors declare no potential conflicts of interest.
References
- 1.Kotsopoulos J, Kim Y-I, Narod SA. Folate and breast cancer: what about high-risk women? Cancer Causes Control 2012;23:1405–20. [DOI] [PubMed] [Google Scholar]
- 2.Strickland KC, Krupenko NI, Krupenko SA. Molecular mechanisms underlying the potentially adverse effects of folate. Clin Chem Lab Med 2013;51:607–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Jacques PF, Selhub J, Bostom AG, et al. The effect of folic acid fortification on plasma folate and total homocysteine concentrations. N Engl J Med 1999;340:1449–54. [DOI] [PubMed] [Google Scholar]
- 4.Lucock M, Yates Z. Folic acid fortification: a double-edged sword. Curr Opin Clin Nutr Metab Care 2009;12:555–64. [DOI] [PubMed] [Google Scholar]
- 5.Choi J-H, Yates Z, Veysey M, et al. Contemporary issues surrounding folic acid fortification initiatives. Prev Nutr Food Sci 2014;19:247–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Herrmann W, Obeid R. The mandatory fortification of staple foods with folic acid. Dtsch Ärztebl Int 2011;108:249–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Mason JB, Levesque T. Folate: effects on carcinogenesis and the potential for cancer chemoprevention. Oncol Williston Park (N.Y.) 1996;10:1727–36. 1742–3; discussion 1743–1744. [PubMed] [Google Scholar]
- 8.Cooper AJ. Biochemistry of sulfur-containing amino acids. Annu Rev Biochem 1983;52:187–222. [DOI] [PubMed] [Google Scholar]
- 9.Scott JM, Weir DG. Folic acid, homocysteine and one-carbon metabolism: a review of the essential biochemistry. J Cardiovasc Risk 1998;5:223–7. [PubMed] [Google Scholar]
- 10.Choi SW. Vitamin B12 deficiency: a new risk factor for breast cancer? Nutr Rev 1999;57:250–3. [DOI] [PubMed] [Google Scholar]
- 11.Duthie SJ, Narayanan S, Brand GM, et al. Impact of folate deficiency on DNA stability. J Nutr 2002; 132:2444S–9S. [DOI] [PubMed] [Google Scholar]
- 12.Blount BC, Mack MM, Wehr CM, et al. Folate deficiency causes uracil misincorporation into human DNA and chromosome breakage: implications for cancer and neuronal damage. Proc Natl Acad Sci USA 1997;94:3290–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Stover PJ. Physiology of folate and vitamin B12 in health and disease. Nutr Rev 2004;62:S3–12. discussion S13. [DOI] [PubMed] [Google Scholar]
- 14.Choi SW, Mason JB. Folate and carcinogenesis: an integrated scheme. J Nutr 2000;130:129–32. [DOI] [PubMed] [Google Scholar]
- 15.Ames BN. DNA damage from micronutrient deficiencies is likely to be a major cause of cancer. Mutat Res 2001;475:7–20. [DOI] [PubMed] [Google Scholar]
- 16.Hansen MF, Jensen SØ, Füchtbauer E-M, et al. High folic acid diet enhances tumour growth in PyMT-induced breast cancer. Br J Cancer 2017; 116:752–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Miller JW, Borowsky AD, Marple TC, et al. Folate, Dna methylation, and mouse models of breast tumorigenesis. Nutr Rev 2008;66:S59–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Lin J, Lee I- M, Cook NR, et al. Plasma folate, vitamin B-6, vitamin B-12, and risk of breast cancer in women. Am J Clin Nutr 2008;87:734–43. [DOI] [PubMed] [Google Scholar]
- 19.Matejcic M, de Batlle J, Ricci C, et al. Biomarkers of folate and vitamin B12 and breast cancer risk: report from the EPIC cohort. Int J Cancer 2017; 140:1246–59. [DOI] [PubMed] [Google Scholar]
- 20.Belanger CF, Hennekens CH, Rosner B, et al. The nurses’ health study. Am J Nurs 1978;78:1039–40. [PubMed] [Google Scholar]
- 21.Colditz GA, Hankinson SE. The nurses’ health study: lifestyle and health among women. Nat Rev Cancer 2005;5:388–96. [DOI] [PubMed] [Google Scholar]
- 22.Bao Y, Bertoia ML, Lenart EB, et al. Origin, methods, and evolution of the three nurses’ health studies. Am J Public Health 2016;106:1573–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Hankinson SE, Willett WC, Manson JE, et al. Alcohol, height, and adiposity in relation to estrogen and prolactin levels in postmenopausal women. J Natl Cancer Inst 1995;87:1297–302. [DOI] [PubMed] [Google Scholar]
- 24.Hankinson SE, Willett WC, Manson JE, et al. Plasma sex steroid hormone levels and risk of breast cancer in postmenopausal women. J Natl Cancer Inst 1998;90:1292–9. [DOI] [PubMed] [Google Scholar]
- 25.Zhang X, Tworoger SS, Eliassen AH, et al. Post-menopausal plasma sex hormone levels and breast cancer risk over 20 years of follow-up. Breast Cancer Res Treat 2013;137:883–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Tamura T, Freeberg LE, Cornwell PE. Inhibition of EDTA of growth of lactobacillus casei in the folate microbiological assay and its reversal by added manganese or iron. Clin Chem 1990;36:1993. [PubMed] [Google Scholar]
- 27.Shin-Buehring YS, Rasshofer R, Endres W. A new enzymatic method for pyridoxal-5-phosphate determination. J Inherit Metab Dis 1981;4:123–4. [Google Scholar]
- 28.Araki A, Sako Y. Determination of free and total homocysteine in human plasma by high-performance liquid chromatography with fluorescence detection. J Chromatogr 1987;422:43–52. [DOI] [PubMed] [Google Scholar]
- 29.Rosner B. Percentage points for a generalized ESD many-outlier procedure. Dent Tech 1983;25:165–72. [Google Scholar]
- 30.Rosner B, Cook N, Portman R, et al. Determination of blood pressure percentiles in normal-weight children: some methodological issues. Am J Epidemiol 2008;167:653–66. [DOI] [PubMed] [Google Scholar]
- 31.Willett WC. Nutritional epidemiology, 3rd ed Oxford, New York: Oxford University Press, 2013. [Google Scholar]
- 32.Tamimi RM, Baer HJ, Marotti J, et al. Comparison of molecular phenotypes of ductal carcinoma in situ and invasive breast cancer. Breast Cancer Res BCR 2008;10:R67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Hirko KA, Chen WY, Willett WC, et al. Alcohol consumption and risk of breast cancer by molecular subtype: prospective analysis of the nurses’ health study after 26 years of follow-up. Int J Cancer 2016;138:1094–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Wang M, Spiegelman D, Kuchiba A, et al. Statistical methods for studying disease subtype heterogeneity. StatMed 2016;35:782–800. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wu K, Helzlsouer KJ, Comstock GW, et al. A prospective study on folate, B12, and pyridoxal 5′-phosphate (B6) and breast cancer. Cancer Epidemiol Biomarkers Prev 1999;8:209–17. [PubMed] [Google Scholar]
- 36.Rossi E, Hung J, Beilby JP, et al. Folate levels and cancer morbidity and mortality: prospective cohort study from Busselton, Western Australia. Ann Epidemiol 2006;16:206–12. [DOI] [PubMed] [Google Scholar]
- 37.Agnoli C, Grioni S, Krogh V, et al. Plasma riboflavin and vitamin B-6, but not Homocysteine, Folate, or vitamin B-12, are inversely associated with breast cancer risk in the European prospective investigation into cancer and nutrition-Varese cohort. J Nutr 2016;146:1227–34. [DOI] [PubMed] [Google Scholar]
- 38.Ericson U, Borgquist S, Ivarsson MIL, et al. Plasma folate concentrations are positively associated with risk of estrogen receptor beta negative breast cancer in a Swedish nested case control study. J Nutr 2010;140:1661–8. [DOI] [PubMed] [Google Scholar]
- 39.Kim SJ, Zuchniak A, Sohn K-J, et al. Plasma folate, vitamin B-6, and vitamin B-12 and breast cancer risk in BRCA1- and BRCA2-mutation carriers: a prospective study. Am J Clin Nutr 2016;104:671–7. [DOI] [PubMed] [Google Scholar]
- 40.Lin J, Lee I-M, Song Y, et al. Plasma homocysteine and cysteine and risk of breast cancer in women. Cancer Res 2010;70:2397–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hazra A, Kraft P, Selhub J, et al. Common variants of FUT2 are associated with plasma vitamin B12 levels. Nat Genet 2008;40:1160–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hazra A, Kraft P, Lazarus R, et al. Genome-wide significant predictors of metabolites in the one-carbon metabolism pathway. Hum Mol Genet 2009;18:4677–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.MacArthur J, Bowler E, Cerezo M, et al. The new NHGRI-EBI catalog of published genome-wide association studies (GWAS catalog). Nucleic Acids Res 2017;45:D896–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zhang SM, Willett WC, Selhub J, et al. Plasma folate, vitamin B6, vitamin B12, homocysteine, and risk of breast cancer. J Natl Cancer Inst 2003; 95:373–80. [DOI] [PubMed] [Google Scholar]
- 45.Zhang SM, Willett WC, Selhub J, et al. A prospective study of plasma total cysteine and risk of breast cancer. Cancer Epidemiol Biomarkers Prev 2003;12:1188–93. [PubMed] [Google Scholar]
- 46.Piyathilake CJ, Macaluso M, Hine RJ, et al. Local and systemic effects of cigarette smoking on folate and vitamin B-12. Am J Clin Nutr 1994;60:559–66. [DOI] [PubMed] [Google Scholar]
- 47.Gabriel HE, Crott JW, Ghandour H, et al. Chronic cigarette smoking is associated with diminished folate status, altered folate form distribution, and increased genetic damage in the buccal mucosa of healthy adults. Am J Clin Nutr 2006;83:835–41. [DOI] [PubMed] [Google Scholar]
- 48.Walmsley CM, Bates CJ, Prentice A, et al. Relationship between cigarette smoking and nutrient intakes and blood status indices of older people living in the UK: further analysis of data from the National Diet and nutrition survey of people aged 65 years and over, 1994/95. Public Health Nutr 1999;2:199–208. [DOI] [PubMed] [Google Scholar]
- 49.Mannino DM, Mulinare J, Ford ES, et al. Tobacco smoke exposure and decreased serum and red blood cell folate levels: data from the Third National Health and Nutrition Examination Survey. Nicotine Tob Res 2003;5:357–62. [DOI] [PubMed] [Google Scholar]
- 50.Jacques PF, Bostom AG, Wilson PW, et al. Determinants of plasma total homocysteine concentration in the Framingham offspring cohort. Am J Clin Nutr 2001;73:613–21. [DOI] [PubMed] [Google Scholar]
- 51.Leenders M, Ros MM, Sluijs I, et al. Reliability of selected antioxidants and compounds involved in one-carbon metabolism in two Dutch cohorts. Nutr Cancer 2013;65:17–24. [DOI] [PubMed] [Google Scholar]
- 52.Midttun Ø, Townsend MK, Nygård O, et al. Most blood biomarkers related to vitamin status, one-carbon metabolism, and the Kynurenine pathway show adequate Preanalytical stability and within-person reproducibility to allow assessment of exposure or nutritional status in healthy women and cardiovascular Patients123. J Nutr 2014;144:784–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Fazili Z, Pfeiffer CM, Zhang M. Comparison of serum folate species analyzed by LC-MS/MS with total folate measured by microbiologic assay and bio-rad radioassay. Clin Chem 2007;53:781–4. [DOI] [PubMed] [Google Scholar]
- 54.Mason JB. Biomarkers of nutrient exposure and status in one-carbon (methyl) metabolism. J Nutr 2003;133(Suppl 3):941S–7S. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.