Abstract
Background:
Tumor infiltrating lymphocytes (TILs) aid in informing treatment for head and neck squamous cell carcinoma (HNSCC). Nevertheless, little is known about the role of diet on TILs.
Methods:
Immunohistologic expression of CD4, CD8, CD68, CD103, CD104 and FOXP3 were assessed in tissue microarrays from 233 previously untreated HNSCC patients. Associations between these markers and pre-treatment dietary patterns were evaluated using linear regression. Associations between baseline serum carotenoids, tocopherols and TILs were assessed using logistic regression. Cox models evaluated the association between diet and TILs on overall and recurrence-free survival.
Results:
Consumption of a Western dietary pattern was associated with lower CD8+ and FOXP3+ infiltrates (p-value:0.03 and 0.02, respectively). Multivariable logistic regression models demonstrated significantly higher CD8+ (OR:2.21;p-value:0.001) and FOXP3+ (OR:4.26;p-value:<0.0001) among patients with high gamma tocopherol. Conversely, high levels of xanthophylls (OR:0.12;p-value:<0.0001), lycopene (OR:0.36;p-value:0.0001) and total carotenoids(OR:0.31;p-value: <0.0001) were associated with significantly lower CD68+. Among those with high CD4+ (HR:1.77;p-value:0.03), CD68+ (HR:2.42;p-value:0.004), CD103+ (HR:3.64;p-value:0.03) and FOXP3+ (HR:3.09;p-value:0.05), having a high Western dietary pattern increased the risk of overall mortality when compared to a low Western dietary pattern.
Conclusion:
Dietary patterns and serum carotenoids may play an important role in modifying TILs, and ultimately, outcome after diagnosis with HNSCC.
Keywords: Head and Neck Cancer, Diet, Epidemiology, Carotenoids, Micronutrients, Serum Markers, Vegetables and Fruit
Introduction
Head and neck cancer (HNC) encompasses malignant lesions found within the epithelial tissue of the oral cavity, pharynx, larynx and nasopharynx, together comprising the sixth most common cancer worldwide (1). More than 90% of neoplasms within the head and neck are reported to be squamous cell carcinoma (HNSCC), commonly associated with extensive exposure to alcohol and tobacco use(2); however, decreases in tobacco consumption in the United States (US)(3, 4) has led to the implication of high-risk human papillomavirus (HPV) as the primary etiologic factor associated with oropharyngeal carcinoma (5). Despite advances in treatment, HNC survival has remained largely stagnant, with global five-year relative survival rates ranging from 40–50% (6). In order to improve patient outcomes, further research and a better understanding of this heterogenous disease is needed.
Recent studies on the role of tumor-infiltrating lymphocytes (TILs) and cancer suggest tumorigenesis to be an immunologic disease as well as genetic one. HNC tumors are unique in that they lie within close proximity to lymphoid tissue in the Waldeyer’s ring, therefore allowing for dense infiltration of immune cells to be easily integrated into the lesion (7). Several studies assessing the role of TILs in HNC have suggested these immune markers to be strong prognostic tools to guide treatment decisions (7). HPV+ HNSCC patients commonly display nodal metastasis, yet the overall and progression-free survival rates are typically better than those among their HPV- counterparts. Due largely to reports of dense lymphocytic infiltrates in HPV+ oropharyngeal carcinoma, one proposed rationale for the observed survival benefit is that the body’s immune response against the viral antigen, particularly in the form of CD8+ and FOXP3+, may aid in driving the elimination of the tumor cells (8–13). TILs have also been repeatedly noted as an important indicator in maximizing treatment efficacy, with increased CD3+, CD4+, CD8+, CD103+ and FOXP3+ T-cell infiltrates correlated with improved survival among HNSCC patients(14–18); conversely, high levels of CD68+ myeloid derived suppressor cells, have previously been reported to be associated with poor HNSCC prognosis(14),
Numerous epidemiologic studies have demonstrated a bi-directional effect of diet on HNSCC risk, progression and prognosis(19–28). While poor nutrition is believed to account for an estimated third of all cancer related deaths in the US (29, 30), the molecular mechanisms underlying the protective effects of bioactive dietary agents remains to be fully elucidated. Immune system modulation through dietary components that can mediate inflammation within the tumor microenvironment is an important mechanism that has been understudied in HNSCC. Although pro-inflammatory factors are essential in the biological response to trauma or infection, excessive production of cytokines, chemokines, growth factors, prostaglandins, and reactive oxygen and nitrogen species have been shown to impact tumor initiation and progression by increasing mutation and cellular proliferation rates, decreasing apoptosis and facilitating angiogenesis (31, 32). Increased consumption of fruits and vegetables have consistently been shown to improve HNSCC prognosis, possibly by counteracting many of the aforementioned inflammatory products through their antioxidant, antimutagenic and antiproliferative properties (26, 33–35). Nevertheless, no studies to date have assessed the role of diet on anti-tumor immune response in HNC.
With increasing interest in the role of tumor immune response in personalized medicine and immunotherapy, it is important to assess the potential role of diet on tumor immune response and, ultimately, outcome. The results of this study will not only impact our knowledge regarding disease pathogenesis, but also aid in facilitating a conversation about dietary recommendations for both HNSCC prevention and intervention.
Methods
Study Population
Incident cases diagnosed with HNSCC were recruited to participate in this prospective cohort study as part of the University of Michigan Head and Neck Specialized Program of Research Excellence (HN-SPORE). Upon approval from the Institutional Review Board at the University of Michigan, study participants provided signed, informed consent to participate. Exclusion criteria included being <18 years of age, pregnant, non-English speaking, diagnosed with another non-upper aerodigestive tract cancer, diagnosed with mental instability, or diagnosed with another head and neck primary within five years preceding the signed consent. In total, 1,042 subjects were enrolled into the study between November 2008 and October 2014.
At baseline, subjects were asked to complete a self-administered health questionnaire that ascertained data on demographic characteristics, tobacco and alcohol use, physical activity, comorbidities, sleep, depression and quality of life. Thirty mL peripheral blood samples were taken prior to treatment using routine venipuncture techniques and formalin-fixed paraffin-embedded (FFPE) tissue blocks were collected from diagnostic biopsies. In order to track patient status, participants were routinely followed every three to six months in clinic. Vital status was reviewed annually following diagnosis. Mortality and recurrence status were obtained by research assistants through medical record review, annual health surveys, the Social Security Death Index and LexisNexis through April 2016. Time to first recurrence was calculated as the number of days between the initial diagnosis at Michigan Medicine and the date of first recurrence. Patients who completed treatment but exhibited persistent disease (were never determined by a medical doctor to be disease-free) were given a recurrence time of 1 day.
For the purpose of this analysis, patients who had not completed their baseline food frequency questionnaire (FFQ) or were not included on a tumor microarray (TMA) were excluded. Additional exclusion criteria included patients who had withdrawn consent, had uncommon tumor sites or unknown primaries, resulting in a final sample size of 233. Finally, for the purposes of the serum carotenoid/tocopherol analysis, those who did not have available baseline serum carotenoid data were excluded, resulting in a sample size of 70.
Specimen and Data Collection
Dietary intake
Dietary intake was collected using the self-administered, semi-quantitative Harvard food frequency questionnaire (FFQ) and analyzed as pretreatment dietary patterns as described previously by Arthur et al.(26). Dietary components and their factor loadings for each of the two dietary patterns can be found in Supplementary Table 2. In brief, the FFQ was designed to evaluate respondents’ typical dietary intake from food and supplements prior to diagnosis. The questionnaire consists of 131 items, and the reproducibility and validity have both been previously reported (36, 37). The FFQ included standard portion sizes for each item (eg: 1 banana, 3–5 oz chicken), which allowed participants to select their average frequency of consumption over the past year from a list of up to 9 choices ranging from “almost never” to “≥6 times per day.” Total energy and nutrient intake were ascertained by summing intakes from each food item based on the selected portion size, reported frequency of consumption, and nutrient content of each food item(36). Estimations for daily food group servings were calculated by summing the frequency weights of each food item based on reported daily frequency of consumption. Upon exclusion based on missing values and energy intake outliers, 39 foods and food groups were isolated using previously defined methods(26, 38, 39). Dietary patterns were deduced by principal component analysis and pattern factor scores were calculated for each patient by summing the reported consumption of the factor food variables weighted by factor loading.
Serum micronutrients
The extraction of serum carotenoids and tocopherols was completed using high-performance liquid chromatography as previously described (40, 41). In summary, equal volumes of serum and ethanol containing butylated hydroxytoluene were mixed and extracted with hexane, utilizing Tocol as the internal standard. To separate carotenoids from tocopherols, a YMC C30 reverse-phase column was used (YMC Company, LTD, Kyoto, Japan) to conduct a gradual elution at 0.2 mL/min total flow on the Shimadzu LC-20AT HPLC system (Shimadzu Corperation, Kyoto, Japan). While detection for carotenoids was at 450 and 472 nm, electrochemical detection using the Coularray electrochemical detector set (Thermo Scientific, Waltham, Mass) was used for Tocol and tocopherols at 310, 390 and 470 mV.
Tumor infiltrating lymphocytes
Hematoxylin-eosin stained slides were created from the aforementioned FFPE tissue specimens and reviewed by an expert pathologist (JM). Upon verification of histology, those slides that expressed >70% cellularity and minimal necrosis were set aside for tissue sampling. Triplicate 0.7mm diameter cores were selected, punched and extracted for each patient to comprise six tissue microarray (TMA) blocks representative of 541 patients. The samples were processed in two batches (Supplemental Table 1)
Slides cut from TMAs were stained for CD4, CD8, CD68, CD103, CD104 and FOXP3 TILs utilizing techniques previously described (14, 17). Briefly, after overnight incubation in a 65°C oven, the sections were deparaffinized, and rehydrated using xylene, graded alcohols, as well as buffer immersion steps. Heat-induced epitope retrieval was then followed by incubation of the slides in a preheated pressure cooker with either Citrate buffer pH6 or Tris-EDTA buffer pH9 and blocked using horse serum. A DAKO autostainer, with chromogens liquid streptavidin biotin horseradish peroxidase and DAB, was then used for immunohistochemical staining. The deparaffinated sections, along with positive and negative controls, were then stained with monoclonal antibodies at the following titrations: CD4–1:250 (Abcam Ab846); CD8–1:40 (Nova Castra VP-C320); CD68 −1:100 (Dako M0814); CD103–1:500 (Abcam Ab129202); CD104 −1:50 (Beta-4 integrin, eBioscience 439–9b); and FoxP3 −1:200 (Abcam Ab20034). The stained slides were digitally imaged to be used in Aperio ImageScope v.12 software (Leica Biosystems).
A technician naive to clinical status scored the stained TMA slides. Grid software (Measure, C Thing Software 2.01) was employed to overlay each tissue core before counting TILs. CD104+ (beta-4 integrin) staining was used before counting TILs to identify the location and extent of carcinoma in each core. Cores were scored as 25%, 50%, 75%, or 100% tumor. Cores deemed to have <25% tumor parenchyma were not scored and therefore excluded from the analysis. Using 200 X magnification, TILs stained with CD4, CD8, CD68, CD103 and FoxP3 antibodies were manually counted. Only those TILs that infiltrated the tumor parenchyma were counted. The TIL count for each core was normalized by dividing each raw value by the fraction of each 0.7 mm core that was indicated as malignant through CD104 staining. Normalizing TIL counts ensured that variation in counted cells was representative of increased TIL density within the tumor parenchyma and not tumor proportion within cores. Normalized mean counts of triplicate samples for each patient were utilized for statistical analysis.
Statistical methods
Descriptive statistics reflective of demographic, clinical and serum carotenoid/tocopherol variables were compared between binary determinants of dietary pattern score. Due to the fact that participants with dietary data were generally healthier than those missing dietary data, we utilized inverse probability weighting (IPW) in order to allow our subcohort patients to better represent the larger HN-SPORE cohort (Supplementary Table 3), thereby improving generalizability(42). Propensity scores (PS) were generated using baseline demographic variables, including: age at diagnosis, sex, stage, disease site, comorbidity score, drinking and smoking status. The reciprocal of the PS was then used as the IPW in statistical analyses. To construct parsimonious statistical models, covariates believed to be relevant to the analyses were chosen by a priori knowledge, input by treating physicians, multicollinearity assessment and determined using backward selection. To assess the association between binary dietary pattern score (around the median) and each continuous TIL marker, both crude and age, sex and TMA adjusted linear regression models were utilized. Due to limited sample size and nonadherence to linear regression assumptions, logistic regression models were used to evaluate associations between TILs and binary serum carotenoids and tocopherols (both individually, and as total groupings); univariate and multivariable models (adjusted for age, sex, disease site and TMA) were compared using Bonferroni correction to account for multiple testing. Finally, Cox proportional hazard (HR) models, adjusted for age, sex, stage, disease site, HPV status and TMA, was used to assess the joint role of dietary pattern and TILs on overall and recurrence-free survival, consequently assessing the potential modification of the relationship between TILs and survival by dietary pattern. All statistical tests were conducted using SAS software 9.4 (SAS Institute, Inc.).
Results
Median follow-up for the 233 participants in this subcohort was 48 months and five year survival was 73.4%; during the course of the study, 65 (27.9%) of patients died and 56 (24.0%) recurred. The average age at diagnosis was 60.5 (SD: 11.3) years. Most of the study subjects were male (73.4%), white (95.7%), and had some post-secondary education (62.5%). The oral cavity was the most common tumor location (44.2%) and just over half of all patients were diagnosed with stage IV tumors (55.8%). Demographic and clinical characteristics stratified by binary dietary pattern score are shown in Table 1a. Individuals who consumed higher quantities of the prudent diet, characterized by high intake of fruits, vegetables, legumes, whole grains and low-fat dairy, were more likely to be older, more highly educated and former smokers, compared to their counterparts, who were more likely to be current smokers. Males were more likely to consume diets high in red and processed meats, refined grains, potatoes, French fries, high-fat dairy, condiments, desserts, snacks and sugar-sweetened beverages characterized by the Western dietary pattern.
Table 1a:
Prudent Diet | Western Pattern | |||
---|---|---|---|---|
Low (N=116) | High (N=117) | Low (N=116) | High (N=117) | |
Age: Mean±SD [range], y | 59±11 [25–95] | 62±11 [39–92] | 62±12 [30–95] | 59±10 [25–92] |
Sex, N (%) | ||||
Male | 89 (79.72) | 82 (70.09) | 77 (66.38) | 94 (80.34) |
Female | 27 (23.28) | 35 (29.91) | 39 (33.62) | 23 (19.66) |
Race | ||||
White | 109 (93.97) | 114 (97.44) | 112 (96.55) | 111 (94.87) |
Black | 3 (2.59) | 1 (0.85) | 1 (0.86) | 3 (2.56) |
American Indian | 3 (2.59) | 1 (0.85) | 1 (0.86) | 3 (2.56) |
Other | 1 (0.86) | 1 (0.85) | 2 (1.72) | 0 (0.00) |
Education | ||||
Less than High School | 10 (8.70) | 6 (5.13) | 9 (7.83) | 7 (5.98) |
High School/GED | 46 (40.00) | 25 (21.37) | 36 (31.30) | 35 (29.91) |
Some College | 42 (36.52) | 45 (38.46) | 40 (34.78) | 47 (40.17) |
4-yr Degree | 10 (8.70) | 15 (12.82) | 11 (9.57) | 14 (11.97) |
More than 4-yr Degree | 7 (6.09) | 26 (22.22) | 19 (16.52) | 14 (40.17) |
BMI: Mean±SD [range], kg/m2 | 26.93±6.02 [16.01–47.94] |
28.15±5.53 [16.44–54.36] |
27.17±5.29 [16.01–40.21] | 27.92±6.26 [16.02–54.36] |
Stage, N (%) | ||||
1 | 16 (13.79) | 19 (16.24) | 18 (15.52) | 17 (14.53) |
2 | 15 (12.93) | 17 (14.53) | 17 (14.66) | 15 (12.82) |
3 | 18 (15.52) | 18 (15.38) | 17 (14.66) | 19 (16.24) |
4 | 67 (57.76) | 63 (53.85) | 64 (55.17) | 66 (56.41) |
Site, N (%) | ||||
Larynx | 21 (18.10) | 22 (18.80) | 14 (12.07) | 29 (24.79) |
Oral Cavity | 50 (43.10) | 53 (45.30) | 55 (47.41) | 48 (41.03) |
Oropharynx | 42 (36.21) | 40 (34.19) | 45 (38.79) | 37 (31.62) |
Hypopharynx | 3 (2.59) | 2 (1.71) | 2 (1.72) | 3 (2.56) |
HPV Status, N (%) * | ||||
Positive | 40 (34.48) | 33 (28.21) | 49 (42.24) | 58 (49.57) |
Negative | 50 (43.10) | 57 (48.72) | 39 (33.62) | 34 (29.06) |
Invalid/Missing | 26 (22.41) | 27 (23.08) | 28 (24.14) | 25 (21.37) |
ACE-27 Score, N (%) | ||||
None | 33 (28.70) | 32 (27.35) | 27 (23.48) | 38 (32.48) |
Mild | 59 (51.30) | 56 (47.86) | 60 (52.17) | 55 (47.01) |
Moderate | 16 (13.91) | 17 (14.53) | 18 (15.65) | 15 (12.82) |
Severe | 7 (6.09) | 12 (10.26) | 10 (8.70) | 9 (7.69) |
Drinking Status, N (%) | ||||
Never | 9 (7.76) | 9 (7.69) | 11 (9.48) | 7 (5.98) |
Current | 80 (68.97) | 84 (71.79) | 75 (64.66) | 89 (76.07) |
Former (quit >12 months) | 27 (23.28) | 24 (20.51) | 30 (25.86) | 21 (17.95) |
Smoking, N (%) | ||||
Never | 30 (25.86) | 27 (23.08) | 36 (31.03) | 21 (17.95) |
Current | 55 (47.41) | 38 (32.48) | 41 (35.34) | 52 (44.44) |
Former (quit >12 months) | 31 (26.72) | 52 (44.44) | 39 (33.62) | 44 (37.61) |
Treatment Modality, N (%) | ||||
Surgery Alone | 28 (24.14) | 30 (25.64) | 29 (25.00) | 29 (24.79) |
Surgery + Adj rad | 17 (14.66) | 19 (16.24) | 16 (13.79) | 20 (17.09) |
Surgery + Adj chemorad | 14 (12.07) | 14 (11.97) | 16 (13.79) | 12 (10.26) |
Radiation Alone | 10 (8.62) | 6 (5.13) | 7 (6.03) | 9 (7.69) |
Chemorad Alone | 38 (32.76) | 38 (32.48) | 40 (34.48) | 36 (30.77) |
Chemo Alone | 1 (0.86) | 3 (2.56) | 1 (0.86) | 3 (2.56) |
Palliative, Unknown | 8 (6.90) | 7 (5.98) | 7 (6.03) | 8 (6.84) |
Deceased, N (%) | 33 (28.45) | 32 (27.35) | 30 (25.86) | 35 (29.91) |
one patient missing ACE-27 score
Abbreviations: ACE-27, Adult Comorbidity Evaluation-27; BMI, body mass index; HPV, human papillomavirus; SD, standard deviation.
Differences in serum carotenoid/tocopherol concentrations by levels of dietary pattern can be found in Table 1b. While beta carotene and total carotenoids were the only biomarkers found to be significantly elevated among high vs. low prudent diet consumers, high Western dietary pattern patients were significantly more likely to have lower levels of beta carotene, xanthophylls, total carotenoids, as well as alpha-, gamma- and total tocopherol when compared to low Western dietary pattern participants.
Table 1b.
Prudent Diet | Western Pattern | |||
---|---|---|---|---|
Low (N=34) | High (N=36) | Low (N=25) | High (N=45) | |
Serum Micronutrients ug/ml, Mean (SD) | ||||
Alpha Carotene | 0.01 (0.02) | 0.02 (0.01) | 0.02 (0.01) | 0.01 (0.02) |
Beta Carotene | 0.05 (0.07) | 0.07 (0.10) | 0.08 (0.11) | 0.04 (0.05) |
Xanthophylls | 0.14 (0.10) | 0.17 (0.13) | 0.18 (0.14) | 0.14 (0.09) |
Lycopene | 0.05 (0.04) | 0.05 (0.04) | 0.05 (0.03) | 0.05 (0.04) |
Total Carotenoids | 0.26 (0.14) | 0.30 (0.24) | 0.34 (0.27) | 0.25 (0.14) |
Alpha tocopherol | 7.98 (5.02) | 10.33 (6.07) | 10.59 (7.49) | 8.34 (4.13) |
Gamma tocopherol | 1.14 (0.57) | 1.18 (0.72) | 1.18 (0.55) | 1.13 (0.79) |
Total tocopherol | 9.12 (5.32) | 11.50 (6.12) | 11.72 (7.76) | 9.52 (4.22) |
Linear regression model results, assessing the association between individual TIL counts and binary dietary pattern (high/low), can be found in Table 2. While no significant associations were found between any of the TILs and the prudent dietary pattern, high intake of the Western dietary pattern was consistently associated with lower levels of CD4+, CD8+, CD103+ and FOXP3+ infiltrates; significant reductions were noted for CD8+ and FOXP3+ markers, even after adjustment for age, sex and TMA (β: −19.99 and −12.54, respectively).
Table 2:
Unadjusted | Adjusted* | |||||||||
---|---|---|---|---|---|---|---|---|---|---|
CD4 | CD8 | CD68 | CD103 | FOXP3 | CD4 | CD8 | CD68 | CD103 | FOXP3 | |
Sample Size | 228 | 228 | 130 | 93 | 232 | 228 | 228 | 130 | 93 | 232 |
Prudent Pattern β(SE) | ||||||||||
≤ −0.20 | ref | ref | ref | ref | ref | ref | ref | ref | ref | ref |
> −0.20 | −1.47 (6.65) | 4.03 (9.13) | 0.86 (4.78) | 19.84 (21.90) | 3.39 (5.37) | −1.89 (6.71) | 3.48 (9.26) | 1.54 (4.92) | 20.82 (22.00) | 4.54 (5.42) |
p-value for trend | 0.82 | 0.66 | 0.86 | 0.37 | 0.53 | 0.78 | 0.71 | 0.76 | 0.35 | 0.40 |
Western Pattern β(SE) | ||||||||||
≤ −0.16 | ref | ref | ref | ref | ref | ref | ref | ref | ref | ref |
> −0.16 | −9.00 (6.63) | −19.99 (9.04) | 0.28 (4.78) | −35.88 (22.00) | −10.78 (5.33) | −8.52 (6.76) | −19.99 (9.27) | −0.13 (4.82) | −41.72 (23.16) | −12.54 (5.42) |
p-value for trend | 0.18 | 0.03 | 0.95 | 0.11 | 0.04 | 0.21 | 0.03 | 0.98 | 0.08 | 0.02 |
Adjusted for age, sex and TMA
Serum carotenoids and tocopherols were evaluated for their association with tumor immune response using logistic regression models and significance was determined based on Bonferroni correction (Table 3). Multivariable models, adjusted for age, sex, disease site and TMA, demonstrated an increased odds of having high CD8+ and FOXP3+ infiltrates among those with high levels of gamma tocopherol (OR: 2.21 [95%CI: 1.37, 3.56] and OR: 4.26 [95%CI: 2.40, 7.58], respectively). High levels of xanthophylls, lycopene, and total carotenoids were shown to decrease the odds of having high CD68+ myeloid derived suppressor cells, a TIL marker previously associated with poor HNSCC prognosis (14) (OR: 0.21 [95%CI: 0.14, 0.32]; OR: 0.53 [95%CI: 0.36, 0.78] and OR: 0.46 [95%CI: 0.31, 0.67], respectively).
Table 3:
Tumor Infiltrating Lymphocytes (TILs) | ||||||||
---|---|---|---|---|---|---|---|---|
Unadjusted | Adjustedǂ | |||||||
CD4 | CD8 | CD68 | FOXP3 | CD4 | CD8 | CD68 | FOXP3 | |
Sample Size | 69 | 69 | 68 | 70 | 69 | 69 | 68 | 70 |
Alpha Carotene | ||||||||
Low (≤ 0.01 ug/ml) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) |
High (>0.01 ug/ml) | 1.48 (1.01, 2.18) | 2.25 (1.53, 3.30) | 0.86 (0.59, 1.25) | 1.54 (1.05, 2.25) | 1.05 (0.64, 1.70) | 1.05 (0.65, 1.71) | 0.53 (0.33, 0.85) | 0.44 (0.25, 0.79) |
p-value | 0.05 | <.0001 | 0.42 | 0.03 | 0.85 | 0.84 | 0.01 | 0.01 |
Beta Carotene | ||||||||
Low (≤0.03 ug/ml) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) |
High (>0.03 ug/ml) | 0.91 (0.62, 1.34) | 1.51 (1.03, 2.20) | 1.08 (0.74, 1.57) | 1.13 (0.78, 1.65) | 0.74 (0.47, 1.17) | 0.95 (0.59, 1.53) | 0.91 (0.58, 1.40) | 0.48 (0.29, 0.82) |
p-value | 0.64 | 0.03 | 0.70 | 0.52 | 0.19 | 0.83 | 0.66 | 0.01 |
Xanthophylls | ||||||||
Low (≤0.13 ug/ml) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) |
High (>0.13 ug/ml) | 1.17 (0.80, 1.71) | 0.98 (0.67, 1.43) | 0.21 (0.14, 0.32) | 0.93 (0.64, 1.36) | 0.73 (0.45, 1.18) | 0.58 (0.35, 0.96) | 0.12 (0.07, 0.21) | 0.91 (0.55, 1.51) |
p-value | 0.43 | 0.92 | <.0001 | 0.72 | 0.20 | 0.03 | <.0001 | 0.71 |
Lycopene | ||||||||
Low (≤0.04 ug/ml) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) |
High (>0.04 ug/ml) | 0.83 (0.56, 1.21) | 1.51 (1.03, 2.20) | 0.53 (0.36, 0.78) | 1.18 (0.81, 1.73) | 0.70 (0.43, 1.14) | 0.47 (0.27, 0.81) | 0.36 (0.22, 0.61) | 1.02 (0.59, 1.75) |
p-value | 0.33 | 0.03 | 0.001 | 0.38 | 0.15 | 0.01 | 0.0001 | 0.95 |
Total* | ||||||||
Low (≤0.24 ug/ml) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) |
High (>0.24 ug/ml) | 0.90 (0.61, 1.32) | 1.23 (0.85, 1.80) | 0.46 (0.31, 0.67) | 0.80 (0.55, 1.17) | 0.61 (0.37, 1.02) | 0.44 (0.26, 0.75) | 0.31 (0.19, 0.51) | 0.56 (0.32, 0.97) |
p-value | 0.57 | 0.27 | <.0001 | 0.25 | 0.06 | 0.003 | <.0001 | 0.04 |
Alpha tocopherol | ||||||||
Low (≤8.42 ug/ml) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) |
High (>8.42 ug/ml) | 2.34 (1.59, 3.46) | 2.09 (1.43, 3.06) | 1.30 (0.89, 1.89) | 1.68 (1.15, 2.44) | 1.61 (1.00, 2.60) | 1.68 (1.04, 2.71) | 0.94 (0.60, 1.47) | 1.08 (0.65, 1.80) |
p-value | <.0001 | 0.0001 | 0.17 | 0.01 | 0.05 | 0.03 | 0.78 | 0.77 |
Gamma tocopherol | ||||||||
Low (≤1.09 ug/ml) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) |
High (>1.09 ug/ml) | 0.69 (0.47, 1.02) | 1.49 (1.02, 2.17) | 1.54 (1.05, 2.24) | 1.45 (1.00, 2.11) | 0.73 (0.46, 1.15) | 2.21 (1.37, 3.56) | 1.89 (1.22, 2.93) | 4.26 (2.40, 7.58) |
p-value | 0.06 | 0.04 | 0.03 | 0.05 | 0.18 | 0.001 | 0.004 | <.0001 |
Total tocopherol ¥ | ||||||||
Low (≤9.44 ug/ml) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) | (Ref) |
High (>9.44 ug/ml) | 1.79 (1.22, 2.64) | 1.60 (1.10, 2.33) | 1.12 (0.77, 1.62) | 1.29 (0.89, 1.88) | 1.25 (0.78, 1.99) | 1.44 (0.90, 2.30) | 0.96 (0.62, 1.49) | 0.93 (0.56, 1.54) |
p-value | 0.003 | 0.01 | 0.56 | 0.18 | 0.35 | 0.13 | 0.86 | 0.78 |
calculates as: alpha carotene+beta carotene+xanthophylls+lycopene
calculated as: alpha tocopherol+gamma tocopherol
Age, sex, disease site and TMA adjusted
Cox proportional hazard model results for the joint association between binary TILs and binary dietary pattern on overall and recurrence free survival, adjusted for age, sex, stage, disease site, HPV status and TMA, can be found in Figures 1 and 2. Surprisingly, among those with high CD4+ TIL counts, having a high prudent dietary pattern (in reference to a low prudent dietary pattern) was associated with increased risk of overall mortality (p for interaction: 0.02). Among those with high CD4+ (p for interaction: 0.03), CD68+ (p for interaction: 0.004), CD103+ (p for interaction: 0.03) and FOXP3+ (p for interaction: 0.05) infiltrates, having a high Western dietary pattern significantly increased the risk of overall mortality when compared to those with a low Western dietary pattern. Similar results were found for recurrence-free survival; among those with high CD4+ (p for interaction: <0.0001) and FOXP3+ (p for interaction 0.0002) TILs, having a high Western dietary pattern appeared to increase the risk of recurrence when compared to those with low Western diet consumption.
Discussion
This study is the first of its kind to report that both pretreatment dietary patterns, as well as serum carotenoids and tocopherols, are not only associated with tumor immune response, but can also modify the association between TILs and survival. Consuming a high Western dietary pattern was consistently associated with lower levels of infiltrates within the tumor, with significant reductions noted for CD8+ and FOXP3+ in multivariable models. Additionally, we found that high serum concentrations of xanthophylls, lycopene and total carotenoids were associated with a lower number of detrimental CD68+ cells in HNSCC tumors, while gamma tocopherol appeared to be associated with significantly higher CD8+ and FOXP3+ lymphocytes. Finally, we demonstrated that the effect the Western dietary pattern exhibits on the tumor microenvironment may have significant negative implications on overall and recurrence-free survival among HNSCC patients.
While numerous studies have analyzed the association between dietary factors and HNSCC (26, 43–46), there have been no studies, to our knowledge, that have assessed the role of dietary patterns or serum carotenoids/tocopherols on tumor immune response at a molecular level. Overall, dietary studies have largely provided evidence that poor nutritional status, often characterized by low consumption of fruits and vegetables, increases the risk of HNSCC(47), while high consumption of nutrient rich foods have the opposite effect(45, 46, 48). Smoking and alcohol consumption, two primary risk factors in HNC development, are believed to play an integral role in counteracting the protective effects conferred by dietary nutrients. Tobacco products, combustible and smokeless, have been shown to produce free radicals and reactive oxygen molecules, which have the ability to react with the lipid bilayer, denature proteins, induce DNA damage, and trigger chronic inflammation (31, 49); high alcohol consumption can lead to decreased folate absorption and increased oxidative stress through ethanol metabolism (50, 51). Nutrients found abundantly in produce have been shown to reduce inflammation and oxidative stress, maintain proper function of cellular processes, and inhibit angiogenesis, theoretically defending high-risk individuals against the carcinogenic effects of tobacco and alcohol use(52, 53). Preclinical and clinical studies have previously elucidated several bioactive agents believed to affect cell-mediated immune response (54, 55). While the prudent diet did not appear to significantly affect TILs in our cohort, we did find several significant associations between serum carotenoids and tocopherols on tumor immune response.
Serum carotenoids and tocopherols have been established by our group and others as valid biomarkers of fruit and vegetable intake (40, 56). Carotenoids, found abundantly in fruits and vegetables, represent a diverse group of natural polyene pigments and can be classified into xanthophylls (b-cryptoxanthin, lutein, and zeaxanthin) and carotenes (a-carotene, b-carotene, and lycopene) (57, 58). Both classes of carotenoids act as antioxidants, efficiently quenching singlet oxygen, reducing damage associated with reactive oxygen species and inhibiting lipid peroxidation (59–61). In this study, we found significantly lower levels of the negative prognostic marker CD68+ associated with higher consumption of xanthophylls, lycopene and total carotenoids. These findings coincide with previous reports that higher pretreatment serum carotenoids are associated with progression-free survival in HNSCC patients(62), as well as the overall survival benefit associated with high serum xanthophyll(35) and lycopene concentrations (63). Tocopherols, commonly known as vitamin E, have long been used clinically for a variety of oxidative stress-induced conditions(64) due to their functionality in trapping free radicals and protecting lipids in the cell membranes from oxidation(65, 66). In our HNC cohort, we found increased levels of serum gamma-tocopherol to significantly increase CD8+ and FOXP3+ infiltrates in adjusted models. Gamma-tocopherol has previously been shown to be a stronger regulator of inflammation than alpha-tocopherol, namely through inhibition of cyclooxygenase (COX) and modulation of reactive oxygen species(67, 68). Several studies have also reported vitamin E supplementation to enhance CD4+ lymphocyte proliferation, and improve CD4+/CD8+ ratios(69, 70). Moreover, gamma-tocopherol, but not alpha-tocopherol, has recently been reported to exhibit anti-proliferative and pro-apoptotic effects selectively on cancer, but not normal epithelial cells (71).
Although limited, several studies have suggested an increased risk of HNSCC incidence and mortality associated with intake of red and processed meat, eggs and dairy (43, 72–74). In this study, the Western dietary pattern, characterized by a high consumption of red and processed meats, refined grains, potatoes, French fries, high-fat dairy, condiments, desserts, snacks and sugar-sweetened beverages, was found to be associated with lower levels of all TILs except CD68+, with significant findings for CD8+ and FOXP3+ in multivariable models. Furthermore, we found that among those individuals who had both dietary and carotenoid data, participants who consumed a high Western dietary pattern had significantly lower levels of beta carotene, xanthophylls, total carotenoids, as well as alpha-, gamma- and total tocopherols. These results suggest that study participants who consumed a high Western dietary pattern were not only consuming unhealthier food but were also eating much fewer fruits and vegetables than those in the low Western dietary pattern group, thereby foregoing the potential protective effects of these nutrients. High intake of red and preserved meats have previously been implicated in other cancers due to the production of genotoxic heterocyclic amines (HCA) and aromatic hydrocarbons (AH) during cooking, as well as nitrates and nitrites for preservation(75–77). The International Agency for Research on Cancer (IARC) has classified the consumption of processed meat as “carcinogenic to humans” (Group 1), and red meat as “probably carcinogenic to humans” (Group 2A) through mechanisms linked to DNA damage and promotion of inflammation (78, 79). Based on the current evidence, it is reasonable to believe that there may be synergistic effects associated with tobacco, alcohol and meat consumption on HNSCC that warrant further investigation.
TILs are of particular importance in HNSCC due to the close proximity of these tumors to lymphoid tissue in the Waldeyer’s ring, as well as the infectious etiology associated with oropharyngeal and nasopharyngeal carcinoma. Albeit being more aggressive, HPV and EBV related malignancies are associated with improved response to treatment, believed to be attributable to the presence of an increased immune response against the viral antigens. Nevertheless, irrespective of infection, TILs have consistently been shown to be an important prognostic tool in guiding HNSCC treatment. In a large retrospective study, increased CD4+ helper T and CD8+ cytotoxic/suppressor T cell levels significantly improved both overall and recurrence free survival in HNC patients; similar findings were reported between these infiltrates and oral cavity(80), laryngeal(81, 82), oro- and hypopharyngeal(12, 13, 16) carcinoma, particularly among patients receiving chemotherapy. FOXP3+ regulatory T cells(14, 83, 84), and more recently CD103+ infiltrates(17), or αEβ7 integrin, have also been shown to be positive prognostic markers in HNSCC, while CD68+ myeloid-derived suppressor cells have been associated with poor prognosis (14). Accounting for the effect of diet on tumor immune response, we found that among those with high CD4+, CD68+, CD103+ and FOXP3+ markers, consumption of a high Western dietary pattern was associated with significantly worse overall survival when compared to those with a low Western dietary pattern; similar findings were noted between high CD4+ and FOXP3+ infiltrates and high Western dietary pattern on recurrence-free survival. These results are particularly interesting because previous studies conducted by our group did not find differential survival among those consuming various levels of the Western diet(26). Given the reduction of all but CD68+ TILs found to be associated with increased Western dietary pattern, and the significant interaction terms between the aforementioned TILs and Western diet, it is reasonable to believe that the protective effect conferred by TILs may be modified by diet. In order to validate these results, future research should be conducted among HNSCC patients.
The results of this study should be interpreted in light of several limitations. Due to the fact that the FFQ was designed to assess dietary intake over the course of the year preceding HNC diagnosis, the effect of lifetime dietary exposure could not be assessed with respect to immune response. In addition, our sample size precluded us from being able to assess the associations of interest in stratified models, particularly evaluating differences in associations by disease site, smoking status or treatment modality. Nevertheless, smoking and alcohol status were not found to be significant confounders when using backward selection, and sensitivity analyses integrating smoking and alcohol consumption into multivariable models did not alter the overall results of the study; treatment modality was found to be highly colinear with disease site, therefore only site was selected as a covariate in adjusted models. While TMAs are significantly more cost and time effective than staining and assessing entire slides, they do not fully capture the heterogeneity present within tumors, thereby limiting our ability to draw conclusions on the overall tumor microenvironment. In an attempt to minimize any potential confounding introduced by using TMAs, we adjusted the TIL counts by the percentage of tumor parenchyma present in each core, ran the samples in triplicate to best assess the overall sample, had a technician blinded to clinical status count the TILs and adjusted for batch effects (by TMA) in statistical models.
In conclusion, dietary patterns, serum carotenoids and tocopherols may play a role in modifying TILs, and ultimately, outcome after diagnosis with HNSCC. As tumor immune response continues to emerge as an important prognostic tool in risk and treatment stratification, particularly with the advent of immunotherapy and personalized medicine, it is important to account for and educate patients on the role of modifiable lifestyle factors in these contexts. While further research is needed in order to confirm these results and elucidate some of the underlying biological mechanisms between Western dietary patterns, inflammation and immune response, the results of this study could inform dietary interventions among high risk individuals in an attempt to curb risk and improve HNSCC prognosis.
Supplementary Material
Funding:
National Institute of Environmental Health Sciences: T32 ES007062
Academy of Nutrition and Dietetics/Colgate Palmolive Fellowship in Nutrition and Oral Health USDA-NIFA Hatch Project 1011487
Footnotes
Conflict of Interest:
None to declare
References:
- 1.Parkin DM, Bray F, Ferlay J, Pisani P: Global cancer statistics, 2002. CA Cancer J Clin 55, 74–108, 2005 [DOI] [PubMed] [Google Scholar]
- 2.Cramer JD, Burtness B, Le QT, Ferris RL: The changing therapeutic landscape of head and neck cancer. Nat Rev Clin Oncol 16, 669–683, 2019. doi: 10.1038/s41571-019-0227-z [DOI] [PubMed] [Google Scholar]
- 3.Alamar B, Glantz SA: Effect of increased social unacceptability of cigarette smoking on reduction in cigarette consumption. American journal of public health 96, 1359–1363, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.CDC: Tobacco use--United States, 1900–1999. MMWR Morbidity and mortality weekly report 48, 986, 1999 [PubMed] [Google Scholar]
- 5.Chaturvedi AK, Engels EA, Pfeiffer RM, Hernandez BY, Xiao W, et al. : Human papillomavirus and rising oropharyngeal cancer incidence in the United States. Journal of clinical oncology 29, 4294–4301, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Head and Neck Cancer: 2014 Review of Cancer Medicines on the WHO List of Essential Medicines.
- 7.Lei Y, Xie Y, Tan YS, Prince ME, Moyer JS, et al. : Telltale tumor infiltrating lymphocytes (TIL) in oral, head & neck cancer. Oral oncology 61, 159–165, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Gunn GR, Zubair A, Peters C, Pan Z-K, Wu T-C, et al. : Two Listeria monocytogenes vaccine vectors that express different molecular forms of human papilloma virus-16 (HPV-16) E7 induce qualitatively different T cell immunity that correlates with their ability to induce regression of established tumors immortalized by HPV-16. The Journal of Immunology 167, 6471–6479, 2001 [DOI] [PubMed] [Google Scholar]
- 9.Psyrri A, Sasaki C, Vassilakopoulou M, Dimitriadis G, Rampias T: Future directions in research, treatment and prevention of HPV-related squamous cell carcinoma of the head and neck. Head and neck pathology 6, 121–128, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Badoual C, Hans S, Merillon N, Van Ryswick C, Ravel P, et al. : PD-1-expressing tumor-infiltrating T cells are a favorable prognostic biomarker in HPV-associated head and neck cancer. Cancer Res 73, 128–38, 2013. doi: 10.1158/0008-5472.CAN-12-2606 [DOI] [PubMed] [Google Scholar]
- 11.Wansom D, Light E, Thomas D, Worden F, Prince M, et al. : Infiltrating lymphocytes and human papillomavirus-16–associated oropharyngeal cancer. The Laryngoscope 122, 121–127, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Nordfors C, Grun N, Tertipis N, Ahrlund-Richter A, Haeggblom L, et al. : CD8+ and CD4+ tumour infiltrating lymphocytes in relation to human papillomavirus status and clinical outcome in tonsillar and base of tongue squamous cell carcinoma. Eur J Cancer 49, 2522–30, 2013. doi: 10.1016/j.ejca.2013.03.019 [DOI] [PubMed] [Google Scholar]
- 13.Nasman A, Romanitan M, Nordfors C, Grun N, Johansson H, et al. : Tumor infiltrating CD8+ and Foxp3+ lymphocytes correlate to clinical outcome and human papillomavirus (HPV) status in tonsillar cancer. PLoS One 7, e38711, 2012. doi: 10.1371/journal.pone.0038711 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Nguyen N, Bellile E, Thomas D, McHug h J, Rozek L, et al. : Tumor infiltrating lymphocytes and survival in patients with head and neck squamous cell carcinoma. Head & neck 38, 1074–1084, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Balermpas P, Michel Y, Wagenblast J, Seitz O, Weiss C, et al. : Tumour-infiltrating lymphocytes predict response to definitive chemoradiotherapy in head and neck cancer. Br J Cancer 110, 501–9, 2014. doi: 10.1038/bjc.2013.640 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Pretscher D, Distel LV, Grabenbauer GG, Wittlinger M, Buettner M, et al. : Distribution of immune cells in head and neck cancer: CD8+ T-cells and CD20+ B-cells in metastatic lymph nodes are associated with favourable outcome in patients with oro- and hypopharyngeal carcinoma. BMC Cancer 9, 292, 2009. doi: 10.1186/1471-2407-9-292 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mann JE, Smith JD, Birkeland AC, Bellile E, Swiecicki P, et al. : Analysis of tumor-infiltrating CD103 resident memory T-cell content in recurrent laryngeal squamous cell carcinoma. Cancer Immunol Immunother 68, 213–220, 2019. doi: 10.1007/s00262-018-2256-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Xiao Y, Li H, Mao L, Yang QC, Fu LQ, et al. : CD103(+) T and Dendritic Cells Indicate a Favorable Prognosis in Oral Cancer. J Dent Res 98, 1480–1487, 2019. doi: 10.1177/0022034519882618 [DOI] [PubMed] [Google Scholar]
- 19.Sandoval M, Font R, Manos M, Dicenta M, Quintana MJ, et al. : The role of vegetable and fruit consumption and other habits on survival following the diagnosis of oral cancer: a prospective study in Spain. International journal of oral and maxillofacial surgery 38, 31–39, 2009 [DOI] [PubMed] [Google Scholar]
- 20.Lucenteforte E, Garavello W, Bosetti C, La Vecchia C: Dietary factors and oral and pharyngeal cancer risk. Oral Oncol 45, 461–7, 2009. doi: 10.1016/j.oraloncology.2008.09.002 [DOI] [PubMed] [Google Scholar]
- 21.Duffy SA, Ronis DL, McLean S, Fowler KE, Gruber SB, et al. : Pretreatment health behaviors predict survival among patients with head and neck squamous cell carcinoma. Journal of clinical oncology 27, 1969–1975, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Li WQ, Park Y, Wu JW, Goldstein AM, Taylor PR, et al. : Index-based dietary patterns and risk of head and neck cancer in a large prospective study. Am J Clin Nutr 99, 559–66, 2014. doi: 10.3945/ajcn.113.073163 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Edefonti V, Bravi F, Garavello W, La Vecchia C, Parpinel M, et al. : Nutrient-based dietary patterns and laryngeal cancer: evidence from an exploratory factor analysis. Cancer Epidemiol Biomarkers Prev 19, 18–27, 2010. doi: 10.1158/1055-9965.EPI-09-0900 [DOI] [PubMed] [Google Scholar]
- 24.Chang CC, Lee WT, Lee YC, Huang CC, Ou CY, et al. : Investigating the association between diet and risk of head and neck cancer in Taiwan. Oncotarget 8, 98865–98875, 2017. doi: 10.18632/oncotarget.22010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Levi F, Pasche C, La Vecchia C, Lucchini F, Franceschi S, et al. : Food groups and risk of oral and pharyngeal cancer. Int J Cancer 77, 705–9, 1998 [DOI] [PubMed] [Google Scholar]
- 26.Arthur AE, Peterson KE, Rozek LS, Taylor JM, Light E, et al. : Pretreatment dietary patterns, weight status, and head and neck squamous cell carcinoma prognosis. The American journal of clinical nutrition 97, 360–368, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Lagiou P, Talamini R, Samoli E, Lagiou A, Ahrens W, et al. : Diet and upper-aerodigestive tract cancer in Europe: the ARCAGE study. Int J Cancer 124, 2671–6, 2009. doi: 10.1002/ijc.24246 [DOI] [PubMed] [Google Scholar]
- 28.Bradshaw PT, Siega-Riz AM, Campbell M, Weissler MC, Funkhouser WK, et al. : Associations between dietary patterns and head and neck cancer: the Carolina head and neck cancer epidemiology study. American journal of epidemiology 175, 1225–1233, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Anand P, Kunnumakara AB, Sundaram C, Harikumar KB, Tharakan ST, et al. : Cancer is a preventable disease that requires major lifestyle changes. Pharmaceutical research 25, 2097–2116, 2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Doll R, Peto R: The causes of cancer: quantitative estimates of avoidable risks of cancer in the United States today. J Natl Cancer Inst 66, 1191–308, 1981 [PubMed] [Google Scholar]
- 31.Grivennikov SI, Greten FR, Karin M: Immunity, inflammation, and cancer. Cell 140, 883–99, 2010. doi: 10.1016/j.cell.2010.01.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Philpott M, Ferguson LR: Immunonutrition and cancer. Mutat Res 551, 29–42, 2004. doi: 10.1016/j.mrfmmm.2004.03.005 [DOI] [PubMed] [Google Scholar]
- 33.Bosetti C, Spertini L, Parpinel M, Gnagnarella P, Lagiou P, et al. : Flavonoids and breast cancer risk in Italy. Cancer Epidemiol Biomarkers Prev 14, 805–8, 2005. doi: 10.1158/1055-9965.EPI-04-0838 [DOI] [PubMed] [Google Scholar]
- 34.Neuhouser ML: Dietary flavonoids and cancer risk: evidence from human population studies. Nutr Cancer 50, 1–7, 2004. doi: 10.1207/s15327914nc5001_1 [DOI] [PubMed] [Google Scholar]
- 35.Arthur AE, Bellile EL, Rozek LS, Peterson KE, Ren J, et al. : Pretreatment serum xanthophyll concentrations as predictors of head and neck cancer recurrence and survival. Head Neck 38 Suppl 1, E1591–7, 2016. doi: 10.1002/hed.24283 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Willett W: Nutritional Epidemiology. 2nd edNew York: Oxford University Press. 1998 [Google Scholar]
- 37.Willett WC, Sampson L, Stampfer MJ, Rosner B, Bain C, et al. : Reproducibility and validity of a semiquantitative food frequency questionnaire. American journal of epidemiology 122, 51–65, 1985 [DOI] [PubMed] [Google Scholar]
- 38.Hu FB, Rimm EB, Stampfer MJ, Ascherio A, Spiegelman D, et al. : Prospective study of major dietary patterns and risk of coronary heart disease in men. Am J Clin Nutr 72, 912–21, 2000. doi: 10.1093/ajcn/72.4.912 [DOI] [PubMed] [Google Scholar]
- 39.Heidemann C, Schulze MB, Franco OH, van Dam RM, Mantzoros CS, et al. : Dietary patterns and risk of mortality from cardiovascular disease, cancer, and all causes in a prospective cohort of women. Circulation 118, 230–7, 2008. doi: 10.1161/CIRCULATIONAHA.108.771881 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Arthur AE, Peterson KE, Shen J, Djuric Z, Taylor JM, et al. : Diet and proinflammatory cytokine levels in head and neck squamous cell carcinoma. Cancer 120, 2704–12, 2014. doi: 10.1002/cncr.28778 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Djuric Z, Ren J, Blythe J, VanLoon G, Sen A: A Mediterranean dietary intervention in healthy American women changes plasma carotenoids and fatty acids in distinct clusters. Nutr Res 29, 156–63, 2009. doi: 10.1016/j.nutres.2009.03.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Brookhart MA, Schneeweiss S, Rothman KJ, Glynn RJ, Avorn J, et al. : Variable selection for propensity score models. Am J Epidemiol 163, 1149–56, 2006. doi: 10.1093/aje/kwj149 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Galeone C, Pelucchi C, Talamini R, Levi F, Bosetti C, et al. : Role of fried foods and oral/pharyngeal and oesophageal cancers. Br J Cancer 92, 2065–9, 2005. doi: 10.1038/sj.bjc.6602542 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Galeone C, Turati F, Zhang ZF, Guercio V, Tavani A, et al. : Relation of allium vegetables intake with head and neck cancers: evidence from the INHANCE consortium. Mol Nutr Food Res 59, 1641–50, 2015. doi: 10.1002/mnfr.201500042 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kreimer AR, Randi G, Herrero R, Castellsague X, La Vecchia C, et al. : Diet and body mass, and oral and oropharyngeal squamous cell carcinomas: analysis from the IARC multinational case-control study. Int J Cancer 118, 2293–7, 2006. doi: 10.1002/ijc.21577 [DOI] [PubMed] [Google Scholar]
- 46.Winn DM: Diet and nutrition in the etiology of oral cancer. Am J Clin Nutr 61, 437S–445S, 1995. doi: 10.1093/ajcn/61.2.437S [DOI] [PubMed] [Google Scholar]
- 47.Gillison ML: Current topics in the epidemiology of oral cavity and oropharyngeal cancers. Head Neck 29, 779–92, 2007. doi: 10.1002/hed.20573 [DOI] [PubMed] [Google Scholar]
- 48.Diet, nutrition, physical activity and cancers of the mouth, pharynx and larynx, 2018. [Google Scholar]
- 49.Takahashi H, Ogata H, Nishigaki R, Broide DH, Karin M: Tobacco smoke promotes lung tumorigenesis by triggering IKKbeta- and JNK1-dependent inflammation. Cancer Cell 17, 89–97, 2010. doi: 10.1016/j.ccr.2009.12.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Pelucchi C, Talamini R, Negri E, Levi F, Conti E, et al. : Folate intake and risk of oral and pharyngeal cancer. Ann Oncol 14, 1677–81, 2003. doi: 10.1093/annonc/mdg448 [DOI] [PubMed] [Google Scholar]
- 51.Poschl G, Seitz HK: Alcohol and cancer. Alcohol Alcohol 39, 155–65, 2004. doi: 10.1093/alcalc/agh057 [DOI] [PubMed] [Google Scholar]
- 52.Hu ML: Dietary polyphenols as antioxidants and anticancer agents: more questions than answers. Chang Gung Med J 34, 449–60, 2011 [PubMed] [Google Scholar]
- 53.Yao H, Xu W, Shi X, Zhang Z: Dietary flavonoids as cancer prevention agents. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 29, 1–31, 2011. doi: 10.1080/10590501.2011.551317 [DOI] [PubMed] [Google Scholar]
- 54.McMurray DN: Cell-mediated immunity in nutritional deficiency. Prog Food Nutr Sci 8, 193–228, 1984 [PubMed] [Google Scholar]
- 55.Janakiram NB, Mohammed A, Madka V, Kumar G, Rao CV: Prevention and treatment of cancers by immune modulating nutrients. Mol Nutr Food Res 60, 1275–94, 2016. doi: 10.1002/mnfr.201500884 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Campbell DR, Gross MD, Martini MC, Grandits GA, Slavin JL, et al. : Plasma carotenoids as biomarkers of vegetable and fruit intake. Cancer Epidemiol Biomarkers Prev 3, 493–500, 1994 [PubMed] [Google Scholar]
- 57.Landrum JT. Carotenoids: physical, chemical, and biological functions and properties: CRC Press, 2009. [Google Scholar]
- 58.Paiva SA, Russell RM: Beta-carotene and other carotenoids as antioxidants. J Am Coll Nutr 18, 426–33, 1999 [DOI] [PubMed] [Google Scholar]
- 59.Fiedor J, Fiedor L, Haessner R, Scheer H: Cyclic endoperoxides of beta-carotene, potential pro-oxidants, as products of chemical quenching of singlet oxygen. Biochim Biophys Acta 1709, 1–4, 2005. doi: 10.1016/j.bbabio.2005.05.008 [DOI] [PubMed] [Google Scholar]
- 60.Cvetkovic D, Fiedor L, Fiedor J, Wiśniewska-Becker A, Markovic D. Molecular base for carotenoids antioxidant activity in model and biological systems: the health-related effects Nova Science Publishers: Hauppauge, NY, USA, 2013. [Google Scholar]
- 61.Krinsky NI, Johnson EJ: Carotenoid actions and their relation to health and disease. Mol Aspects Med 26, 459–516, 2005. doi: 10.1016/j.mam.2005.10.001 [DOI] [PubMed] [Google Scholar]
- 62.Sakhi AK, Russnes KM, Thoresen M, Bastani NE, Karlsen A, et al. : Pre-radiotherapy plasma carotenoids and markers of oxidative stress are associated with survival in head and neck squamous cell carcinoma patients: a prospective study. BMC Cancer 9, 458, 2009. doi: 10.1186/1471-2407-9-458 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Mayne ST, Cartmel B, Lin H, Zheng T, Goodwin WJ Jr: Low plasma lycopene concentration is associated with increased mortality in a cohort of patients with prior oral, pharynx or larynx cancers. Journal of the American College of Nutrition 23, 34–42, 2004 [DOI] [PubMed] [Google Scholar]
- 64.Bieri JG, Corash L, Hubbard VS: Medical uses of vitamin E. N Engl J Med 308, 1063–71, 1983. doi: 10.1056/NEJM198305053081805 [DOI] [PubMed] [Google Scholar]
- 65.Seidman MD, Quirk WS, Shirwany NA: Reactive oxygen metabolites, antioxidants and head and neck cancer. Head Neck 21, 467–79, 1999 [DOI] [PubMed] [Google Scholar]
- 66.Schorah CJ: Micronutrients, antioxidants and risk of cancer. Bibl Nutr Dieta, 92–107, 1995. [DOI] [PubMed] [Google Scholar]
- 67.Dietrich M, Traber MG, Jacques PF, Cross CE, Hu Y, et al. : Does gamma-tocopherol play a role in the primary prevention of heart disease and cancer? A review. J Am Coll Nutr 25, 292–9, 2006 [DOI] [PubMed] [Google Scholar]
- 68.Jiang Q, Christen S, Shigenaga MK, Ames BN: gamma-tocopherol, the major form of vitamin E in the US diet, deserves more attention. Am J Clin Nutr 74, 714–22, 2001. doi: 10.1093/ajcn/74.6.714 [DOI] [PubMed] [Google Scholar]
- 69.Lee CY, Man-Fan Wan J: Vitamin E supplementation improves cell-mediated immunity and oxidative stress of Asian men and women. J Nutr 130, 2932–7, 2000. doi: 10.1093/jn/130.12.2932 [DOI] [PubMed] [Google Scholar]
- 70.Chavance M, Brubacher G, Herbeth B, Vernhes G, Mikstacki T, et al. : Immunological and nutritional status among the elderly. Nutritional Immunity and Illne8s in the Elderly Elm8ford, NY: Pergamon Press Inc, 137–142, 1985 [Google Scholar]
- 71.Jiang Q, Wong J, Fyrst H, Saba JD, Ames BN: gamma-Tocopherol or combinations of vitamin E forms induce cell death in human prostate cancer cells by interrupting sphingolipid synthesis. Proc Natl Acad Sci U S A 101, 17825–30, 2004. doi: 10.1073/pnas.0408340102 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Launoy G, Milan C, Day NE, Pienkowski MP, Gignoux M, et al. : Diet and squamous-cell cancer of the oesophagus: a French multicentre case-control study. Int J Cancer 76, 7–12, 1998 [DOI] [PubMed] [Google Scholar]
- 73.Bosetti C, Talamini R, Levi F, Negri E, Franceschi S, et al. : Fried foods: a risk factor for laryngeal cancer? Br J Cancer 87, 1230–3, 2002. doi: 10.1038/sj.bjc.6600639 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Toporcov TN, Antunes JL, Tavares MR: Fat food habitual intake and risk of oral cancer. Oral Oncol 40, 925–31, 2004. doi: 10.1016/j.oraloncology.2004.04.007 [DOI] [PubMed] [Google Scholar]
- 75.Zheng W, Lee SA: Well-done meat intake, heterocyclic amine exposure, and cancer risk. Nutr Cancer 61, 437–46, 2009. doi: 10.1080/01635580802710741 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Pan A, Sun Q, Bernstein AM, Schulze MB, Manson JE, et al. : Red meat consumption and mortality: results from 2 prospective cohort studies. Arch Intern Med 172, 555–63, 2012. doi: 10.1001/archinternmed.2011.2287 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ferguson LR: Meat and cancer. Meat Sci 84, 308–13, 2010. doi: 10.1016/j.meatsci.2009.06.032 [DOI] [PubMed] [Google Scholar]
- 78.Bouvard V, Loomis D, Guyton KZ, Grosse Y, El Ghissassi F, et al. : Carcinogenicity of consumption of red and processed meat. The Lancet Oncology 16, 1599–1600, 2015 [DOI] [PubMed] [Google Scholar]
- 79.Research WCRFAIfC. Diet, Nutrition, Physical Activity and Cancer: a Global Perspective, 2018. [Google Scholar]
- 80.Wolf GT, Chepeha DB, Bellile E, Nguyen A, Thomas D, et al. : Tumor infiltrating lymphocytes (TIL) and prognosis in oral cavity squamous carcinoma: a preliminary study. Oral Oncol 51, 90–5, 2015. doi: 10.1016/j.oraloncology.2014.09.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hoesli R, Birkeland AC, Rosko AJ, Issa M, Chow KL, et al. : Proportion of CD4 and CD8 tumor infiltrating lymphocytes predicts survival in persistent/recurrent laryngeal squamous cell carcinoma. Oral Oncol 77, 83–89, 2018. doi: 10.1016/j.oraloncology.2017.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Dewyer NA, Wolf GT, Light E, Worden F, Urba S, et al. : Circulating CD4-positive lymphocyte levels as predictor of response to induction chemotherapy in patients with advanced laryngeal cancer. Head Neck 36, 9–14, 2014. doi: 10.1002/hed.23263 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Bron L, Jandus C, Andrejevic-Blant S, Speiser DE, Monnier P, et al. : Prognostic value of arginase-II expression and regulatory T-cell infiltration in head and neck squamous cell carcinoma. Int J Cancer 132, E85–93, 2013. doi: 10.1002/ijc.27728 [DOI] [PubMed] [Google Scholar]
- 84.Badoual C, Hans S, Rodriguez J, Peyrard S, Klein C, et al. : Prognostic value of tumor-infiltrating CD4+ T-cell subpopulations in head and neck cancers. Clin Cancer Res 12, 465–72, 2006. doi: 10.1158/1078-0432.CCR-05-1886 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.