Article Text

Original article
Plasma 25-hydroxyvitamin D and colorectal cancer risk according to tumour immunity status
  1. Mingyang Song1,2,
  2. Reiko Nishihara1,3,
  3. Molin Wang2,
  4. Andrew T Chan4,5,
  5. Zhi Rong Qian3,
  6. Kentaro Inamura3,6,
  7. Xuehong Zhang5,
  8. Kimmie Ng3,
  9. Sun A Kim3,
  10. Kosuke Mima3,
  11. Yasutaka Sukawa3,
  12. Katsuhiko Nosho7,
  13. Charles S Fuchs3,5,
  14. Edward L Giovannucci1,2,5,
  15. Kana Wu1,
  16. Shuji Ogino2,3,5,8
  1. 1Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
  2. 2Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts, USA
  3. 3Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts, USA
  4. 4Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts, USA
  5. 5Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA
  6. 6Laboratory of Human Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
  7. 7Department of Gastroenterology, Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Japan
  8. 8Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts, USA
  1. Correspondence to Dr Shuji Ogino, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, 450 Brookline Ave, Room M422, Boston, MA 02215, USA; shuji_ogino{at}dfci.harvard.edu

Abstract

Objective Evidence suggests protective effects of vitamin D and antitumour immunity on colorectal cancer risk. Immune cells in tumour microenvironment can convert 25-hydroxyvitamin D [25(OH)D] to bioactive 1α,25-dihydroxyvitamin D3, which influences neoplastic and immune cells as an autocrine and paracrine factor. Thus, we hypothesised that the inverse association between vitamin D and colorectal cancer risk might be stronger for cancers with high-level immune response than those with low-level immune response.

Design We designed a nested case–control study (318 rectal and colon carcinoma cases and 624 matched controls) within the Nurses’ Health Study and Health Professionals Follow-up Study using molecular pathological epidemiology database. Multivariable conditional logistic regression was used to assess the association of plasma 25(OH)D with tumour subtypes according to the degree of lymphocytic reaction, tumour-infiltrating T cells (CD3+, CD8+, CD45RO+ (PTPRC) and FOXP3+ cells), microsatellite instability or CpG island methylator phenotype.

Results The association of plasma 25(OH)D with colorectal carcinoma differed by the degree of intratumoural periglandular reaction (p for heterogeneity=0.001); high 25(OH)D was associated with lower risk of tumour with high-level reaction (comparing the highest versus lowest tertile: OR 0.10; 95% CI 0.03 to 0.35; p for trend<0.001), but not risk of tumour with lower-level reaction (p for trend>0.50). A statistically non-significant difference was observed for the associations of 25(OH)D with tumour subtypes according to CD3+ T cell density (p for heterogeneity=0.03; adjusted statistical significance level of α=0.006).

Conclusions High plasma 25(OH)D level is associated with lower risk of colorectal cancer with intense immune reaction, supporting a role of vitamin D in cancer immunoprevention through tumour–host interaction.

  • COLORECTAL CANCER
  • EPIDEMIOLOGY
  • NUTRITION
  • IMMUNOLOGY
  • IMMUNOTHERAPY

Statistics from Altmetric.com

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

Significance of this study

What is already known on this subject?

  • Vitamin D has been associated with lower risk of colorectal cancer (CRC).

  • Vitamin D plays an important role in regulation of immune function.

  • The multifaceted roles of host immunity and inflammation in regulating tumour evolution have long been recognised.

What are the new findings?

  • The association of high-level plasma vitamin D and lower risk of CRC differs by CRC subtypes classified by the degree of lymphocytic reaction to CRC.

  • Plasma vitamin D level is associated with lower risk of the CRC subtype characterised by high-degree intratumoural periglandular reaction, but not risk of the CRC subtype with low-degree reaction.

How might it impact on clinical practice in the foreseeable future?

  • Our findings provide the first line of population-based evidence for a role of vitamin D in cancer immunoprevention through tumour–host interaction.

  • In the future, host immunity status may serve as a potential biomarker to predict the benefit from vitamin D supplementation or other vitamin D-augmenting intervention for CRC prevention.

Introduction

Colorectal cancer (CRC) is the third most common cancer and the fourth cause of cancer death worldwide. Studies have shown that high level of circulating vitamin D is associated with lower CRC risk,1 supporting a preventive effect of vitamin D against CRC.2 Obtained from food, supplements or photochemical synthesis in the skin, vitamin D is hydroxylated in the liver to the major circulating form, 25-hydroxyvitamin D [25(OH)D], and further hydroxylated to the biologically active form of vitamin D, 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3], by some specific cells in the body including immune cells.3–5

Accumulating evidence indicates an important role of vitamin D in regulation of immune function.6 ,7 Likewise, the multifaceted roles of host immunity and inflammation in regulating tumour evolution have long been recognised.8–12 Local immunity status in tumour microenvironment may eliminate transformed cells or promote their tumourigenic potential, thus determining the fate of emerging tumour.13 However, despite compelling evidence for the role of vitamin D in immunity and the role of immunity in tumour development, no study has yet examined whether the inverse association between vitamin D and CRC risk differs according to CRC subtypes classified by immunity status in the tumour microenvironment. When assessing cancer immunity, it is important to examine immune cells in the tumour microenvironment, which exhibit a substantial phenotypic difference from the same immune cell type in peripheral blood.14 We speculated that immune cells in the tumour microenvironment might augment the antitumour effect of vitamin D by means of their ability to enzymatically convert 25(OH)D to 1,25(OH)2D3. Therefore, we hypothesised that the lower CRC risk associated with high-level plasma vitamin D might be stronger for CRC subtype characterised by high-level immune cell infiltrates than for other CRC subtype with low-level immune cell infiltrates.

To test this hypothesis, we investigated the association of plasma 25(OH)D levels with risk of CRC subtypes according to the pattern and intensity of lymphocytic reaction to CRC in a nested case–control study within two large prospective cohort studies, the Nurses’ Health Study (NHS) and the Health Professionals Follow-up Study (HPFS). We additionally examined densities of tumour-infiltrating T cell subsets. Higher levels of lymphocytic reaction to CRC and tumour-infiltrating T cells have been strongly associated with survival of patients with CRC independent of tumour molecular features in these two cohorts.15 ,16 The two cohort studies offered us a unique opportunity to integrate data on prediagnostic plasma vitamin D level and immune cell evaluation in CRC tissue specimens in the longitudinal follow-up scheme. This integrative approach has enabled us to provide novel population-based evidence for possible interactive roles of vitamin D and host immunity in CRC prevention.

Methods

Study population

The NHS enrolled 121 701 registered female nurses in the USA who were aged 30–55 years at baseline in 1976, and the HPFS included 51 529 USA male professionals who were aged 40–75 years at baseline in 1986.17 In both cohorts, follow-up questionnaires were administered at baseline and biennially thereafter to collect and update medical, lifestyle and other health-related information; validated food frequency questionnaires were completed every 4 years to update dietary information. More details about the two cohorts can be found in the online supplementary materials.

In both cohorts, when participants reported a diagnosis of colon or rectal carcinoma in biennial questionnaires, we asked for permission to acquire their medical records and pathological reports. We identified deaths, including lethal unreported CRC cases, through the National Death Index and next of kin. For CRC deaths, we requested permission from next of kin to review medical records. A study physician, blinded to 25(OH)D information, reviewed records to confirm CRC diagnosis and extract relevant information on anatomic location, stage and histological type of the cancer.

Blood specimens were collected from 32 826 women in the NHS between 1989 and 1990; and from 18 225 men in the HPFS between 1993 and 1995. The procedures for blood collection, handling and storage were similar for the two cohorts, as previously described.18 Among participants who provided plasma samples, we documented 400 incident CRC cases in the NHS during follow-up through 1 June 2010 and 299 CRC cases in the HPFS through 31 January 2010. We collected paraffin-embedded archival tissue blocks from hospitals where participants with CRC had undergone tumour resection. For the current study, to minimise the influence of subclinical emerging tumour on plasma 25(OH)D level, we excluded CRC cases that were diagnosed within 2 years after blood draw. We also excluded cases if their plasma samples failed in 25(OH)D measurement or tumour lymphocytic reaction could not be determined. For each case, we used risk set sampling to randomly select up to two controls matched on sex (cohort), age (within 2 years) and year/month of blood draw (within 1 month in the same year) from eligible participants who were alive and free of cancer (except for non-melanoma skin cancer) at the time of diagnosis of the CRC case. As a result, 172 CRC cases and 342 controls from the NHS, and 146 cases and 282 controls from the HPFS were included in the analysis (figure 1).

Figure 1

Flow diagram of the nested case–control study design within the Nurses’ Health Study (NHS) and the Health Professionals Follow-up Study (HPFS). 25(OH)D, 25-hydroxyvitamin D; CRC, colorectal cancer.

Plasma 25(OH)D assay

Plasma 25(OH)D was measured using a radioimmunosorbent assay at the laboratory of Dr Bruce Hollis (Medical University of South Carolina, Charleston, South Carolina, USA) and Heartland Assays as described elsewhere.18 ,19 Samples from cases and their matched controls were handled together and analysed in the same batch. Quality control samples were randomly interspersed among the case–control samples. Personnel blinded to quality control and case–control status conducted all assays. The mean intra-assay coefficient of variation from quality control samples was <15% for all batches.

Plasma inflammatory marker assays

To account for the potential confounding effect by systemic inflammation on the plasma 25(OH)D-CRC association, we also measured three inflammatory markers in our study samples: C-reactive protein (CRP), interleukin-6 (IL6) and tumour necrosis factor receptor superfamily member 1B (TNFRSF1B, also known as soluble tumour necrosis factor receptor 2). We used a highly sensitive immunoturbidimetric assay (Denka Seiken Co, Tokyo, Japan) to measure CRP levels, an ultrasensitive ELISA (R&D Systems, Minneapolis, Minnesota, USA) to measure IL6 and an ELISA (R&D Systems) to measure TNFRSF1B levels. More details regarding the measurements can be found in previous publications.20 ,21

Tumour immunity and molecular analyses

A pathologist (SO) evaluated tissue sections of patients with CRC for the four components of lymphocytic reaction, including intratumoural periglandular reaction, tumour-infiltrating lymphocytes, Crohn’s-like lymphoid reaction and peritumoural lymphocytic reaction.16 Each component was evaluated as absent, mild, moderate or marked, and an agreement study was conducted as previously described.16 In the current analyses, we combined moderate and marked lymphocytic infiltrate categories (as ‘high’) because of low-case counts in these categories.

We also constructed tissue microarray22 to assess the density of tumour-infiltrating CD3+, CD8+, CD45RO+ (PTPRC) and FOXP3+ T cells. We used immunohistochemistry techniques, an automated scanning microscope and Ariol image analysis system (Genetix, San Jose, California, USA) to calculate the average density (cells/mm2) of each T cell subset in tissue microarray cores, as previously described.15 We dichotomised cases based on the density of each T cell subset using the cut-off given in the footnote of table 3.

Because lymphocytic reaction to CRC has been associated with microsatellite instability (MSI) and CpG island methylator phenotype (CIMP) in CRC,16 we also assessed MSI and CIMP status using the DNA extracted from tissue specimens as previously described.23–25 More details are provided in the online supplementary materials.

Statistical analysis

Details of the statistical analysis are provided in the online supplementary materials. We used SAS V.9.3 for all analyses (SAS Institute Inc, Cary, North Carolina, USA). All statistical tests were two sided. Our primary hypothesis testing was the heterogeneity test between ‘the association of plasma 25(OH)D with lymphocyte-rich CRC subtype’ and ‘that with lymphocyte-poor CRC subtype’. To account for multiple testing for the eight primary hypotheses associated with the eight immunity variables (degrees of intratumoural periglandular reaction, tumour-infiltrating lymphocytes, Crohn’s-like reaction and peritumoural reaction; and densities of CD3+, CD8+, CD45RO+ and FOXP3+ T cells), we corrected the statistical significance level to α=0.05/8=0.006 by the Bonferroni correction. All other assessments including evaluation of individual OR estimates represented our secondary analyses. We recognised the use of multiple comparisons and interpreted our data cautiously.

Plasma 25(OH)D levels were categorised into tertiles within each batch of measurement on the basis of the distribution among controls. We used multivariable conditional logistic regression to estimate ORs for CRC subtypes in relation to tertiles of plasma 25(OH)D. Test for trend was performed using the median value for each tertile as a continuous variable in the regression models. To examine the heterogeneity in the associations with various CRC subtypes, we used likelihood ratio test with one degree of freedom by comparing the model in which the association with plasma 25(OH)D was allowed to vary by tumour subtypes (ordinal or binary) to a model in which a common association was assumed across tumour subtypes.22 ,26

We tested whether plasma 25(OH)D-CRC association varied by cohort using the Q statistic before pooling.27 The association was similar in the two cohorts (comparing extreme tertiles: multivariable OR 0.71; 95% CI 0.50 to 1.01; p for trend=0.05 in NHS; OR 0.79; 95% CI 0.52 to 1.21; p for trend=0.29 in HPFS), and no statistically significant difference was detected (p for heterogeneity=0.66 for Cochran's Q test). Therefore, for our main analyses, we pooled data from both cohorts.

Results

Baseline characteristics of study participants

Table 1 shows basic characteristics of our study population. When compared with controls, CRC cases tended to be obese and smoke before age 30 in men (p=0.02). In contrast, compared with cases, controls tended to take aspirin regularly and consume less alcohol, more folate and calcium in women (p<0.05). The median of plasma 25(OH)D concentrations was higher among controls (27.8 ng/mL in women, 29.2 ng/mL in men) than cases (26.1 ng/mL in women, 27.8 ng/mL in men) (p=0.02 in women, p=0.10 in men).

Table 1

Age-adjusted basic characteristics of case and control participants in women (1990) and men (1994)*

By comparing the baseline characteristics of cases with and without tumour immunity data, we did not find any substantial difference between the two groups except for lower alcohol consumption and higher proportion of stage II and III tumours among cases that had lymphocytic reaction data than those without lymphocyte data in women (see online supplementary table S1).

We observed that plasma 25(OH)D level was not significantly associated with lower risk of overall CRC in our nested case–control set (p for trend=0.09; table 2). Such association did not appreciably differ by the availability of tumour immunity data (p for heterogeneity>0.60) when the CRC cases in the current study were compared with the patients with CRC who were excluded due to the unavailable tumour immunity data (see online supplementary table S2).

Table 2

Plasma 25-hydroxyvitamin D levels and colorectal cancer, overall and by components of lymphocytic reaction*

Plasma 25(OH)D level and CRC subtypes classified by degrees of lymphocytic reactions and densities of T cell subsets

We examined degrees of lymphocytic reaction in tissue sections of CRC. Table 2 shows the association of plasma 25(OH)D level with risk of CRC subtypes classified by the degrees of lymphocytic reactions. Our primary hypothesis testing was on heterogeneity between ‘the association of plasma 25(OH)D with risk of lymphocyte-rich CRC’ and ‘that with risk of lymphocyte-poor CRC’ in the combined cohort, and the statistical significance level was adjusted to α=0.006 to account for multiple testing. Notably, the association of plasma 25(OH)D with risk of CRC subtypes differed by the degree of intratumoural periglandular reaction (p for heterogeneity=0.001).

High plasma 25(OH)D level was statistically significantly associated with lower risk of CRC subtype possessing high-level intratumoural periglandular reaction (comparing the highest versus the lowest tertiles: multivariable OR 0.10; 95% CI 0.03 to 0.35; p for trend<0.001 across tertiles of 25(OH)D level), but not with CRC subtypes possessing absent or mild reaction (p for trend=0.93 for the mild-reaction subtype; and p for trend=0.55 for the absent-reaction subtype). A similar but attenuated difference was observed between risks of CRC subtypes classified by tumour-infiltrating lymphocytes, Crohn's-like reaction or peritumoural reaction; and the heterogeneity test did not reach statistical significance (p for heterogeneity>0.07).

We additionally subclassified CRC according to densities of each of the four T cell subsets (CD3+, CD8+, CD45RO+ and FOXP3+ cells) within CRC tissues (table 3). Similar to the results on intratumoural periglandular reaction, the association between plasma 25(OH)D level and CRC risk differed by CD3+ T cell density (p for heterogeneity=0.03), although this difference was not significant at the stringent statistical significance level (α=0.006). High level of plasma 25(OH)D was associated with lower risk of colorectal tumours that were infiltrated by high density of CD3+ cells (p for trend=0.006), but not with tumours having low density of CD3+ cells (p for trend=0.77). The association of plasma 25(OH)D with risk of CRC did not significantly differ by the density of CD8+, CD45RO+ or FOXP3+ cells (p for heterogeneity>0.10).

Table 3

Plasma 25-hydroxyvitamin D levels and colorectal cancer, overall and by tumour-infiltrating T-cell subset density*

Plasma 25(OH)D level and CRC subtypes classified by MSI or CIMP status

Because lymphocytic reaction to CRC has been associated with MSI and CIMP in CRC,16 we also classified tumours by MSI and CIMP status as our secondary analyses. The association of plasma 25(OH)D with CRC subtypes did not significantly differ by MSI (p for heterogeneity=0.02) or CIMP status (p for heterogeneity=0.76; online supplementary table S3) at the stringent statistical significance level (α=0.006).

Sensitivity analysis

To further control for potential confounding by lifestyle factors, instead of adjusting for the Dietary Approaches to Stop Hypertension (DASH) score, we adjusted for individual dietary factors that have been related to CRC risk, including multivitamins, calcium, red meat and processed meat, and total fibre, in our multivariable model. The results remained very similar, and the p value for heterogeneity was 0.002 for the associations between plasma 25(OH)D and risk of CRC subtypes classified by intratumoural periglandular reaction (see online supplementary table S4). Given the potential influence of systemic inflammation on plasma 25(OH)D status and CRC development,28 ,29 we also adjusted for quartiles of each of the three inflammatory markers (ie, CRP, IL6 and TNFRSF1B) in the multivariable model. As shown in online supplementary table S5, the results did not essentially change and the plasma 25(OH)D-CRC associations remained statistically significantly different according to the degree of intratumoural periglandular reaction (p for heterogeneity<0.001).

Discussion

We conducted this study to test the hypothesis that the inverse association of plasma vitamin D level with risk of CRC might be stronger for CRC subtype with high-level lymphocytic reaction than for CRC subtype with low-level reaction. We found that the relationship between plasma 25(OH)D and risk of CRC differed by intratumoural periglandular reaction to CRC; high 25(OH)D was associated with lower risk of tumours possessing high-level lymphocytic reaction, but not with tumours having low-level or no lymphocytic reaction. Although statistical significance was not reached at the stringent level (α=0.006), we also observed that the inverse association of plasma 25(OH)D with CRC risk appeared to be stronger for tumours infiltrated with high density of CD3+ T cells than for tumours with lower density of CD3+ T cells. Our data provide evidence for a possible role of tumour stromal immune cells in generating bioactive 1,25(OH)2D3 to augment the influence of vitamin D on neoplastic and non-neoplastic cells in an autocrine and paracrine fashion.

As cancer immunotherapy has become an attractive strategy, integrated analyses of tumour molecular features and host factors including dietary and environmental exposures and immune response to tumour are increasingly important.30–33 The degree of lymphocytic infiltrate in CRC tissue has been associated with MSI status and better patient survival.34–38 However, there is a paucity of data on epidemiological exposures (such as plasma 25(OH)D level) combined with tumour molecular features and immune response in the tumour microenvironment in population-based studies. Our current study aimed to address this challenge.

Although numerous epidemiological studies have shown a lower CRC risk associated with high vitamin D level, it is still of considerable debate about whether this represents a causal relationship or arises from confounding. In a large randomised trial, daily supplementation with 400 IU of vitamin D combined with 1000 mg of elemental calcium for 7 years had no detectable benefit for CRC occurrence.39 However, methodological limitations of this trial, including inadequate vitamin D dose, duration and compliance, might have contributed to the null findings. In this context, investigation of the influence of vitamin D on CRC subtypes characterised by immunity-related pathological features may not only provide important insight into the causality of the vitamin D–CRC relationship but also reveal a complex interaction between exposures, host factors and tumour cells.40 Our findings provide the first line of population-based evidence for the role of host immunity in vitamin D-mediated CRC prevention, therefore generating some mechanistic hypotheses for further investigation.

One possible mechanism through which immune response may modulate the effect of vitamin D on carcinogenesis is that immune cells can convert 25(OH)D to bioactive 1,25(OH)2D3, and thereby enhancing the effect of vitamin D on behaviours of both neoplastic and non-neoplastic cells. Macrophages, dendritic cells, T cells and B cells have all been shown to express enzymes critical for vitamin D metabolism and have an immune autocrine/paracrine activity.3–5 Locally synthesised 1,25(OH)2D3 can then bind to the vitamin D receptor (VDR) and regulate transcription of genes that control cell proliferation, apoptosis and differentiation.2 In contrast, colorectal neoplasia with low lymphocytic infiltrates may not have sufficient bioactive vitamin D in the tumour microenvironment to mediate the influence of plasma 25(OH)D level on neoplasia evolution.

As an alternative mechanism, modulation of immune function by vitamin D may help maintain immune homeostasis of the intestine, thus favouring tumour-suppressive effects over tumour-promoting effect of lymphocytic infiltrates.7 It is plausible that the tumour-suppressive effect of vitamin D may be more pronounced in emerging tumours with abundant immune cells than in those with fewer immune cells. The role of inflammation caused by gut microbiota or other stimuli in colorectal carcinogenesis has been increasingly recognised.41 ,42 Vitamin D exerts an inhibitory action on the adaptive immune system through suppressing proinflammatory TH1 cell activity43 and enhancing anti-inflammatory TH2 cell activity.44 There is evidence suggesting that adequate vitamin D and VDR expression are required for T cell antigen receptor signalling and subsequent T cell activation.45 Recently, vitamin D has been found to prevent inflammation-associated colon cancer through suppression of inflammatory responses during initiation of carcinogenesis.46 These experimental data may be consistent with our observation of the strong inverse association of high plasma 25(OH)D with risk of CRC subtype with high-level lymphocytic reactions.

Our current study has limitations. First, the sample size is limited due to the necessity of both prediagnostic plasma and CRC tissue specimens, and therefore our results should be interpreted cautiously. Given the uniqueness of the current study, our findings need to be replicated in independent datasets. Second, potential selection bias might arise from our exclusion of CRC cases without available tumour tissue data. However, the distribution of risk factors among included cases did not appreciably differ from excluded cases. Moreover, the association between plasma 25(OH)D and risk of overall CRC did not appreciably differ by the availability (versus unavailability) of tumour tissue data. Third, the study was observational and subject to influence of confounding. However, adjustment for a wide range of risk factors for CRC had minimal impact on our results.

Our study has several strengths. First, this longitudinal study was conducted within two well-defined cohorts, and our nested case–control design enabled us to match each CRC case with controls from the same background population, which represents a substantial advantage over ordinary case–control design. Second, we measured 25(OH)D in plasma specimens obtained from cohort participants when they had not known if they would develop CRC or not in the future. These prediagnostic plasma specimens represent a precious resource to evaluate plasma biomarkers for a potential risk assessment tool in clinical settings. Third, we collected detailed information on potential confounders and had a high follow-up rate of the cohorts. Fourth, this study represents a unique integrative molecular pathological epidemiology47–52 analysis of prediagnostic plasma vitamin D and immunity status in tumour tissue, which has enabled us to provide novel epidemiological evidence on the potential role of vitamin D in cancer immunoprevention.

In conclusion, high-level plasma 25(OH)D is associated with a lower risk of the CRC subtype characterised by intense intratumoural periglandular lymphocytic reaction, but not with risk of CRC subtypes with less intense reaction. Our findings suggest a potential interplay of vitamin D and immune system that may operate to prevent CRC development. Further research is needed to confirm our findings and to examine potential mechanisms for CRC immunoprevention.

Acknowledgments

We deeply thank hospitals and pathology departments throughout the USA for generously providing us with tissue specimens. We would also like to thank the participants and staff of the Nurses’ Health Study and the Health Professionals Follow-up Study for their valuable contributions as well as the following state cancer registries for their help: AL, AZ, AR, CA, CO, CT, DE, FL, GA, ID, IL, IN, IA, KY, LA, ME, MD, MA, MI, NE, NH, NJ, NY, NC, ND, OH, OK, OR, PA, RI, SC, TN, TX, VA, WA and WY. We assume full responsibility for analyses and interpretation of these data.

References

Supplementary materials

  • Supplementary Data

    This web only file has been produced by the BMJ Publishing Group from an electronic file supplied by the author(s) and has not been edited for content.

    Files in this Data Supplement:

Footnotes

  • MS, RN, MW, ATC, CSF, ELG, KW and SO contributed equally. We use Human Genome Organisation (HUGO) Gene Nomenclature Committee (HGNC)-approved symbols for genes and gene products, including CD3, CD8, CRP, FOXP3, IL6, PTPRC, TNFRSF1B, and VDR; all of which are described at http://www.genenames.org

  • Contributors MS and SO have full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis. Study concept and design: MS, ELG, KW and SO. Acquisition of data: MS, RN, MW, ATC, ZRQ, KI, XZ, KN, SAK, KM, YS, KN, CSF, ELG, KW and SO. Analysis and interpretation of data: MS, RN, MW, ATC, ZRQ, KI, XZ, KN, SAK, KM, YS, KN, CSF, ELG, KW and SO. Drafting of the manuscript: MS and SO. Critical revision of the manuscript for important intellectual content: MS, ATC, XZ, ELG, KW and SO. Statistical analysis: MS, RN, MW and SO. Funding acquisition: ATC, CSF, ELG and SO. Administrative, technical or material support: ATC, CSF, ELG and SO. Study supervision: SO.

  • Funding This work was supported by US National Institutes of Health (NIH) grants [P01 CA87969 to SE Hankinson; R01 CA49449 to SE Hankinson; UM1 CA186107 to MJ Stampfer; P01 CA55075 to WC Willett; UM1 CA167552 to WC Willett; R03 CA176717 to XZ; R01 CA137178 to ATC; K24 DK098311 to ATC; P50 CA127003 to CSF; DF/HCC GI SPORE Developmental Project to SO; R01 CA151993 to SO; K07 CA190673 to RN; and K07 CA148894 to KN], and grants from The Paula and Russell Agrusa Fund for Colorectal Cancer Research (to CSF), the Friends of the Dana-Farber Cancer Institute (SO), the Bennett Family Fund, and the Entertainment Industry Foundation. ATC is a Damon Runyon Clinical Investigator. MS is a trainee of the Harvard Transdisciplinary Research Center on Energetics and Cancer (TREC). KI is supported by a Japan Society for the Promotion of Science Postdoctoral Fellowship for Research Abroad and by Takashi Tsuruo Memorial Fund. SAK is supported by an early exchange postdoctoral fellowship grant from Asian medical centre.

  • Competing interests ATC previously served as a consultant for Bayer Healthcare, Millennium Pharmaceuticals, and Pfizer Inc. This study was not funded by Bayer Healthcare, Millennium Pharmaceuticals or Pfizer.

  • Patient consent Obtained.

  • Ethics approval The Brigham and Women's Hospital and the Harvard School of Public Health.

  • Provenance and peer review Not commissioned; externally peer reviewed.