Background: Lung cancer is the leading cause of cancer death. Little is known about whether prediagnostic nutritional factors may affect survival. We examined the associations of prediagnostic calcium intake from foods and/or supplements with lung cancer survival.

Methods: The present analysis included 23,882 incident, primary lung cancer patients from 12 prospective cohort studies. Dietary calcium intake was assessed using food-frequency questionnaires at baseline in each cohort and standardized to caloric intake of 2,000 kcal/d for women and 2,500 kcal/d for men. Stratified, multivariable-adjusted Cox regression was applied to compute hazard ratios (HR) and 95% confidence intervals (CI).

Results: The 5-year survival rates were 56%, 21%, and 5.7% for localized, regional, and distant stage lung cancer, respectively. Low prediagnostic dietary calcium intake (<500–600 mg/d, less than half of the recommendation) was associated with a small increase in risk of death compared with recommended calcium intakes (800–1,200 mg/d); HR (95% CI) was 1.07 (1.01–1.13) after adjusting for age, stage, histology, grade, smoking status, pack-years, and other potential prognostic factors. The association between low calcium intake and higher lung cancer mortality was evident primarily among localized/regional stage patients, with HR (95% CI) of 1.15 (1.04–1.27). No association was found for supplemental calcium with survival in the multivariable-adjusted model.

Conclusions: This large pooled analysis is the first, to our knowledge, to indicate that low prediagnostic dietary calcium intake may be associated with poorer survival among early-stage lung cancer patients.

Impact: This multinational prospective study linked low calcium intake to lung cancer prognosis. Cancer Epidemiol Biomarkers Prev; 26(7); 1060–70. ©2017 AACR.

Lung cancer is the leading cause of cancer death worldwide, accounting for 1.6 million deaths (20% of all cancer deaths) annually (1). Most lung cancer patients are diagnosed at advanced stages; the 5-year survival rate is only ∼18% in the United States and is even lower in other countries (2–4). While its prognosis largely depends on stage, histologic type, treatment options, and patient demographics and comorbidity status (2, 5, 6), emerging evidence suggests that prediagnostic nutrition and lifestyle factors may also influence lung cancer survival (7–9).

Both experimental and epidemiological studies have suggested potential roles of calcium in cancer development and progression (10). Besides its well-known effects on bone health, calcium intake and calcium homeostasis can affect cell proliferation, differentiation, and apoptosis, parathyroid hormone (PTH) and PTH-related peptide, vitamin D metabolism and signaling, angiogenesis, and immune response (11, 12). Compromised bone health and impaired calcium homeostasis due to a long-term calcium insufficiency may promote tumor progression and metastasis. Cohort studies and meta-analyses have linked adequate calcium intake with decreased overall cancer risk and risks of colorectal, breast, and prostate cancers (13–19). Evidence, although limited, has also linked calcium intake with lung cancer risk (20–22). However, few cohort studies have examined the association of prediagnostic calcium intake with cancer survival, and none explicitly with lung cancer survival (23, 24).

We investigated prediagnostic calcium intake from foods and supplements in relation to lung cancer survival using data from a large pooling project, the Calcium and Lung Cancer Pooling Project. Individual-level data of ∼1.7 million participants from 12 prospective cohort studies in the United States, Europe, and Asia were pooled, resulting in 23,882 incident, primary lung cancer cases. We examined the association among all cases combined and separately by major lung cancer prognostic factors, including age, stage, and histology. We focused our analyses more on the survival of localized and regional stage patients because it is unlikely that the prognosis of distant stage lung cancer would be significantly influenced by prediagnostic nutritional factors, given its high fatality rate and short survival time.

Study populations

Participating studies included eight US cohorts: the National Institutes of Health–AARP study (NIH-AARP; ref. 25), Health Professionals' Follow-Up Study (HPFS; ref. 26), Nurses' Health Study I (NHS; ref. 27), Iowa Women's Health Study (IWHS; ref. 28), Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial (PLCO; ref. 29), Southern Community Cohort Study (SCCS; ref. 30), Vitamins and Lifestyle Cohort Study (VITAL; ref. 31), and Women's Health Initiative Observational Study (WHI; ref. 32); one European cohort: the European Prospective Investigation into Cancer and Nutrition Cohort (EPIC; ref. 33); and three Asian cohorts: the Japan Public Health Center-based Prospective Study cohort I and II (JPHC; ref. 34), Shanghai Men's Health Study (SMHS; ref. 35), and Shanghai Women's Health Study (SWHS; ref. 36). Each study was approved by the Institutional Review Board at local institutions; the pooling project was approved by the Vanderbilt University Institutional Review Board.

Assessment of dietary and supplemental calcium intake

Usual dietary intakes were assessed at baseline in each cohort using a self- or interviewer-administered food-frequency questionnaire (FFQ) as described in previous publications (37–48). The FFQs usually inquired about habitual consumption of common food items over the past 12 months and were validated against 24-hour dietary recalls, 7-day food records, or dietary biomarkers. Daily food intakes were estimated based on the frequency and amount of consumption and were linked to country/region-specific food composition tables to calculate intakes of energy (kcal/d), calcium (mg/d), and other nutrients. In the current study, dietary intakes were adjusted for total energy intake using the nutrient density method (49) and standardized to intakes per 2,000 kcal for women and per 2,500 kcal for men. Intake of supplemental calcium was assessed in eight US cohorts. Participants were asked whether in the past year they generally took supplements (multivitamins and/or single calcium supplements); and if yes, how often (from less than once per week to every day) and how much they usually took (from <200 mg/d to >1,000 mg/d for calcium). Most cohorts estimated supplemental calcium intakes from both calcium supplements and multivitamins, except that the SCCS asked only about the use of calcium supplements.

Assessment of lung cancer incidence and survival

Our primary outcome is overall survival time, calculated by counting from the date of lung cancer diagnosis to the date of death or end of follow-up, whichever came first. Incident cancer cases and the vital status of cancer patients were identified in each cohort through linkages with national/regional cancer registries and death registries, follow-up interviews, review of medical records and/or death certificates, or these methods combined. Cancers of the bronchus and lung were ascertained by the International Classification of Diseases (ICD) codes: 162 (ICD-9) or C34 (ICD-10). Year of diagnosis and whether lung cancer was the underlying cause of death was acquired from the cohorts. Clinical information was obtained when available, including stage, histologic type, and grade. We harmonized tumor information across studies as follows: stage—localized, regional, distant, and unknown; histologic type—adenocarcinoma, squamous cell carcinoma, other non–small cell lung cancer, small cell lung cancer, and other; and grade—well-, moderately, and poorly differentiated, undifferentiated, and unknown.

Assessment of non-dietary covariates

Baseline information on sociodemographics, lifestyles, medical history, and anthropometrics was obtained from each cohort and harmonized for the current study, including age at baseline and at diagnosis (years, integer), sex (male or female), race/ethnicity (non-Hispanic white, Black, Asian, or other), educational attainment (≤high school, vocational school or some college, college or graduate school), smoking status (never, former, or current use of cigarettes, cigars, or pipe), pack-years of cigarette smoking (continuous), alcohol drinking status [none, moderate, or heavy (>14 g/d for women and >28 g/d for men)], physical activity level [low, middle, or high (cutoffs: zero leisure-time physical activity and median of non-zero leisure-time physical activity assessed by metabolic equivalents or hours, or tertile of total physical activity metabolic equivalents)], history of diabetes (yes or no), obesity status (body mass index [BMI] <18.5, 18.5–24.99, 25.0–29.99, or ≥30 kg/m2), and in women, postmenopausal status (yes or no) and use of hormone therapy (never or ever).

The proportion of missing values was generally <10% in each cohort for these covariates. If the proportion of missing values was <3%, we assigned the median nonmissing value for continuous variables (e.g., BMI) and the most frequent category for categorical variables (e.g., education). If the proportion of missing variables was ≥3%, we used a multivariate imputation to estimate missing value based on other covariates, calcium intake, energy intake, and lung cancer and death outcomes (SAS PROC MI procedure). Missing data imputation was processed for each cohort separately. Of note, in the JPHC, Cohort I did not have data on physical activity metabolic equivalents and Cohort II did not collect information on education level; we imputed these two variables using the above described method in JPHC Cohort I and II data combined.

Analytic population

We excluded participants from the current study if they had (i) a history of any cancer except non-melanoma skin cancer prior to lung cancer, (ii) missing diagnosis or survival time information, (iii) missing calcium intakes or smoking status information, or (iv) implausible total energy intake (beyond three standard deviations of the cohort- and sex-specific log-transformed mean energy intake or beyond the cohort predetermined range). A total of 24,440 incident primary lung cancer cases among 1,679,842 participants of the pooling project were considered eligible. We further excluded 11 cases with cancer in situ and 547 cases with data missing on both stage and histology, leaving 23,882 incident lung cancer cases in our analyses.

Statistical analysis

Usual dietary calcium intakes were compared among lung cancer patients with different baseline characteristics and tumor features using a general linear model (adjusted for age at baseline, sex, and total energy intake). Corresponding 5-year survival rates were estimated by the life-table method and P for differences was evaluated via the log-rank test with Bonferroni correction. The Cox proportional hazard model was used to estimate hazard ratios (HR) and 95% confidence intervals (CI), stratified by cohort, year of lung cancer diagnosis (5-year intervals from earlier than 1990 to later than 2010), and time interval between dietary assessment and lung cancer diagnosis (<4, 4–7, >7–10, and >10 years, according to the quartile distribution). Potential confounders associated with calcium intake and/or lung cancer survival were adjusted for, including age at diagnosis, sex, race/ethnicity, education, smoking status, pack-years of cigarette smoking, total energy intake, alcohol consumption, physical activity, history of diabetes, obesity status, use of hormone therapy in women, and the stage, histologic type, and grade of lung cancer. Considering the interplay of calcium, magnesium, vitamin D, and phosphorus, we further adjusted for, when data were available, dietary intakes of magnesium (in all cohorts), vitamin D (in 9 cohorts), and phosphorus (in all cohorts), individually or together, with or without interaction terms with calcium. However, the associations of dietary calcium with lung cancer survival were basically unchanged, so these nutrients were not included in the final model.

Calcium intakes were analyzed as categorical variables and as continuous variables. We used the Dietary Reference Intakes recommended by the US Institute of Medicine as project-wide cutoff points for dietary and total calcium intakes (11). Briefly, for men ages 19 to 70 years and women ages 19 to 50 years, the estimated average requirement (EAR) of calcium is 800 mg/d and the recommended dietary allowance (RDA) is 1,000 mg/d; for men >70 years and women >50 years, the EAR is 1,000 mg/d and the RDA is 1,200 mg/d. Participants were classified into five groups based on their calcium intakes: <0.5 RDA, 0.5 RDA to EAR, EAR to RDA, RDA to 1.5 RDA, or >1.5 RDA. The cutoff points for supplemental calcium intake were 0, 200, 500, and 1,000 mg/d. A joint analysis was conducted by dietary and supplemental calcium intakes to examine specifically the association for supplemental calcium among those with low dietary calcium intake and the association for dietary calcium among those with no or little supplemental calcium intake. Calcium intakes were modeled continuously in restricted cubic spline regression to examine dose–response associations. Patients in the sex-specific top and bottom 1% of calcium intakes were excluded. Three knots were chosen based on model fitness, at the 5th, 50th, and 95th percentiles (corresponding to 425, 910, and 1625 mg/d). The referent intake was 900 mg/d, and all potential confounders were included in the spline regression.

Stratified analyses were performed by potential effect modifiers, including age at diagnosis, sex, race/ethnicity, education, smoking, other lifestyle factors, stage, histologic type, grade, and time interval between dietary assessment and cancer diagnosis. P for interaction was evaluated via likelihood ratio test comparing models with and without the interaction term (calcium intake category × stratification variable). Sensitivity analyses were conducted by excluding those diagnosed with lung cancer within two years after baseline, by excluding those who died or were lost to follow-up within 3 months after diagnoses, or by examining lung cancer-specific mortality. Meta-analysis was applied as an alternative approach to pooled analysis. Cohort-specific HRs and 95% CI were calculated and then combined using a fixed-effect model because no significant between-study heterogeneity was detected. Two-sided P < 0.05 were considered statistically significant. All analyses were conducted using SAS, version 9.4 (SAS Institute, Inc.).

Among 1,679,842 men and women from 12 cohort studies, 23,882 incident primary lung cancer cases were identified during a median follow-up of 7 years (interquartile range, 4–10 years); 19,538 died (16,279 from lung cancer) with a median survival time of 11 months (interquartile range, 4–34 months). The overall 5-year survival rate was 21.3%. Higher survival rates were associated with younger age at diagnosis, female gender, never smoking, fewer pack-years if ever smoked, no history of diabetes, and higher level of physical activity (Table 1). Particularly low 5-year survival rates were found for small cell lung cancer (9.7%), distant stage cancer (5.7%), and undifferentiated tumor cells (10.4%). Dietary calcium intakes were higher in the US and European cohorts than in Asian cohorts (Supplementary Table S1) and were positively associated with past smoking, physical activity, moderate alcohol consumption, BMI, history of diabetes, and use of hormone therapy in women. Prediagnostic dietary calcium intakes did not vary by tumor characteristics.

Table 1.

Characteristics, dietary calcium intake, and 5-year survival rate of lung cancer cases (n = 23,882)

Characteristics Cases, n Deaths, n Dietary calcium intake, mg/da P for calcium intakeb 5-Year survival rate (95% CI), % P for survival rateb
Age at diagnosis 
 <65 years  6,439  5,087  883 ± 413  ref  23.8 (22.8–24.9)  ref 
 65–75 years  13,263  11,039  930 ± 412  <0.0001  21.0 (20.3–21.7)  <0.0001 
 >75 years  4,180  3,412  947 ± 412  <0.0001  18.0 (16.8–19.2)  <0.0001 
Sex 
 Female  11,574  9,021  897 ± 425  ref  25.3 (24.5–26.2)  ref 
 Male  12,308  10,517  944 ± 424  <0.0001  17.5 (16.8–18.2)  <0.0001 
Race 
 White  19,448  16,137  979 ± 389  ref  21.1 (20.6–21.7)  ref 
 Black  1,037  780  789 ± 392  <0.0001  22.5 (19.9–25.2)  0.99 
 Asian  3,140  2,408  593 ± 396  <0.0001  21.8 (20.3–23.3)  0.76 
 Other  257  213  924 ± 388  0.14  20.0 (15.3–25.2)  0.99 
Education 
 High school  9,947  8,181  889 ± 409  ref  18.8 (18.0–19.6)  ref 
 Vocational school/Some college  8,140  6,668  963 ± 410  <0.0001  22.2 (21.2–23.1)  <0.0001 
 College and graduate school  5,795  4,689  912 ± 414  0.002  23.6 (22.5–24.6)  <0.0001 
Tobacco smoking 
 Never  2,948  2,063  867 ± 415  ref  31.6 (29.8–33.3)  ref 
 Former  9,589  7,972  975 ± 420  <0.0001  21.4 (20.6–22.2)  <0.0001 
 Current  11,345  9,503  889 ± 415  0.04  18.6 (17.8–19.3)  <0.0001 
Smoking pack-years in cigarette smokers 
 <30 pack-years  6,187  4,876  946 ± 412  ref  22.8 (21.7–23.9)  ref 
 30–50 pack-years  7,442  6,275  917 ± 410  <0.0001  19.4 (18.5–20.3)  <0.0001 
 >50 pack-years  7,134  6,173  923 ± 418  0.005  18.0 (17.1–18.9)  <0.0001 
History of diabetes 
 No  22,028  17,918  913 ± 409  ref  21.8 (21.2–22.3)  ref 
 Yes  1,854  1,620  1007 ± 411  <0.0001  15.5 (13.9–17.2)  <0.0001 
Physical activity 
 Low  5,674  4,565  836 ± 407  ref  20.0 (18.9–21.0)  ref 
 Middle  9,157  7,631  924 ± 407  <0.0001  20.7 (19.9–21.6)  0.22 
 High  9,051  7,342  969 ± 406  <0.0001  22.6 (21.7–23.5)  <0.0001 
Body mass index 
 <18.5 kg/m2  525  423  796 ± 408  0.0002  21.7 (20.9–22.5)  0.35 
 18.5–24.99 kg/m2  10,758  8,768  889 ± 410  ref  20.3 (16.9–24.0)  ref 
 25.0–29.99 kg/m2  8,888  7,294  948 ± 411  <0.0001  21.0 (20.1–21.9)  0.86 
 ≥30.0 kg/m2  3,711  3,053  963 ± 408  <0.0001  20.8 (19.5–22.2)  0.68 
Alcohol consumption 
 None  7,265  5,849  909 ± 404  ref  21.4 (20.5–22.4)  ref 
 Moderate  11,414  9,341  989 ± 404  <0.0001  21.9 (21.2–22.7)  0.98 
 Heavy  5,203  4,348  786 ± 411  <0.0001  19.5 (18.5–20.6)  0.02 
Hormone therapy among women 
 No  6,686  5,263  867 ± 388  ref  24.1 (23.1–25.2)  ref 
 Yes  4,888  3,758  931 ± 388  <0.0001  27.0 (25.7–28.3)  <0.0001 
Histological type 
 Adenocarcinoma  9,621  7,324  922 ± 411  ref  27.7 (26.8–28.7)  ref 
 Squamous cell carcinoma  4,318  3,468  913 ± 411  0.99  24.7 (23.4–26.0)  <0.0001 
 Other non–small cell carcinoma  3,414  2,769  958 ± 409  0.001  21.6 (20.2–23.0)  <0.0001 
 Small cell carcinoma  3,210  2,955  945 ± 409  0.05  9.7 (8.7–10.8)  <0.0001 
 All other  3,319  3,022  861 ± 410  <0.0001  9.6 (8.7–10.7)  <0.0001 
Tumor stage 
 Localized  3,489  1,823  921 ± 410  ref  56.4 (54.6–58.1)  ref 
 Regional  4,524  3,713  905 ± 410  0.54  21.1 (20.0–22.3)  <0.0001 
 Distant  6,752  6,327  904 ± 412  0.31  5.7 (5.2–6.3)  <0.0001 
 Unknown  9,117  7,675  940 ± 416  0.12  20.4 (19.5–21.2)  <0.0001 
Tumor grade 
 Well differentiated  1,213  592  843 ± 409  ref  57.9 (54.9–60.8)  ref 
 Moderately differentiated  2,791  1,953  921 ± 409  <0.0001  37.3 (35.5–39.1)  0.66 
 Poorly differentiated  4,560  3,742  925 ± 411  <0.0001  22.2 (21.0–23.4)  <0.0001 
 Undifferentiated  1,838  1,714  831 ± 409  0.99  10.4 (9.0–11.8)  <0.0001 
 Unknown  13,480  11,537  938 ± 410  <0.0001  16.2 (15.5–16.8)  <0.0001 
Characteristics Cases, n Deaths, n Dietary calcium intake, mg/da P for calcium intakeb 5-Year survival rate (95% CI), % P for survival rateb
Age at diagnosis 
 <65 years  6,439  5,087  883 ± 413  ref  23.8 (22.8–24.9)  ref 
 65–75 years  13,263  11,039  930 ± 412  <0.0001  21.0 (20.3–21.7)  <0.0001 
 >75 years  4,180  3,412  947 ± 412  <0.0001  18.0 (16.8–19.2)  <0.0001 
Sex 
 Female  11,574  9,021  897 ± 425  ref  25.3 (24.5–26.2)  ref 
 Male  12,308  10,517  944 ± 424  <0.0001  17.5 (16.8–18.2)  <0.0001 
Race 
 White  19,448  16,137  979 ± 389  ref  21.1 (20.6–21.7)  ref 
 Black  1,037  780  789 ± 392  <0.0001  22.5 (19.9–25.2)  0.99 
 Asian  3,140  2,408  593 ± 396  <0.0001  21.8 (20.3–23.3)  0.76 
 Other  257  213  924 ± 388  0.14  20.0 (15.3–25.2)  0.99 
Education 
 High school  9,947  8,181  889 ± 409  ref  18.8 (18.0–19.6)  ref 
 Vocational school/Some college  8,140  6,668  963 ± 410  <0.0001  22.2 (21.2–23.1)  <0.0001 
 College and graduate school  5,795  4,689  912 ± 414  0.002  23.6 (22.5–24.6)  <0.0001 
Tobacco smoking 
 Never  2,948  2,063  867 ± 415  ref  31.6 (29.8–33.3)  ref 
 Former  9,589  7,972  975 ± 420  <0.0001  21.4 (20.6–22.2)  <0.0001 
 Current  11,345  9,503  889 ± 415  0.04  18.6 (17.8–19.3)  <0.0001 
Smoking pack-years in cigarette smokers 
 <30 pack-years  6,187  4,876  946 ± 412  ref  22.8 (21.7–23.9)  ref 
 30–50 pack-years  7,442  6,275  917 ± 410  <0.0001  19.4 (18.5–20.3)  <0.0001 
 >50 pack-years  7,134  6,173  923 ± 418  0.005  18.0 (17.1–18.9)  <0.0001 
History of diabetes 
 No  22,028  17,918  913 ± 409  ref  21.8 (21.2–22.3)  ref 
 Yes  1,854  1,620  1007 ± 411  <0.0001  15.5 (13.9–17.2)  <0.0001 
Physical activity 
 Low  5,674  4,565  836 ± 407  ref  20.0 (18.9–21.0)  ref 
 Middle  9,157  7,631  924 ± 407  <0.0001  20.7 (19.9–21.6)  0.22 
 High  9,051  7,342  969 ± 406  <0.0001  22.6 (21.7–23.5)  <0.0001 
Body mass index 
 <18.5 kg/m2  525  423  796 ± 408  0.0002  21.7 (20.9–22.5)  0.35 
 18.5–24.99 kg/m2  10,758  8,768  889 ± 410  ref  20.3 (16.9–24.0)  ref 
 25.0–29.99 kg/m2  8,888  7,294  948 ± 411  <0.0001  21.0 (20.1–21.9)  0.86 
 ≥30.0 kg/m2  3,711  3,053  963 ± 408  <0.0001  20.8 (19.5–22.2)  0.68 
Alcohol consumption 
 None  7,265  5,849  909 ± 404  ref  21.4 (20.5–22.4)  ref 
 Moderate  11,414  9,341  989 ± 404  <0.0001  21.9 (21.2–22.7)  0.98 
 Heavy  5,203  4,348  786 ± 411  <0.0001  19.5 (18.5–20.6)  0.02 
Hormone therapy among women 
 No  6,686  5,263  867 ± 388  ref  24.1 (23.1–25.2)  ref 
 Yes  4,888  3,758  931 ± 388  <0.0001  27.0 (25.7–28.3)  <0.0001 
Histological type 
 Adenocarcinoma  9,621  7,324  922 ± 411  ref  27.7 (26.8–28.7)  ref 
 Squamous cell carcinoma  4,318  3,468  913 ± 411  0.99  24.7 (23.4–26.0)  <0.0001 
 Other non–small cell carcinoma  3,414  2,769  958 ± 409  0.001  21.6 (20.2–23.0)  <0.0001 
 Small cell carcinoma  3,210  2,955  945 ± 409  0.05  9.7 (8.7–10.8)  <0.0001 
 All other  3,319  3,022  861 ± 410  <0.0001  9.6 (8.7–10.7)  <0.0001 
Tumor stage 
 Localized  3,489  1,823  921 ± 410  ref  56.4 (54.6–58.1)  ref 
 Regional  4,524  3,713  905 ± 410  0.54  21.1 (20.0–22.3)  <0.0001 
 Distant  6,752  6,327  904 ± 412  0.31  5.7 (5.2–6.3)  <0.0001 
 Unknown  9,117  7,675  940 ± 416  0.12  20.4 (19.5–21.2)  <0.0001 
Tumor grade 
 Well differentiated  1,213  592  843 ± 409  ref  57.9 (54.9–60.8)  ref 
 Moderately differentiated  2,791  1,953  921 ± 409  <0.0001  37.3 (35.5–39.1)  0.66 
 Poorly differentiated  4,560  3,742  925 ± 411  <0.0001  22.2 (21.0–23.4)  <0.0001 
 Undifferentiated  1,838  1,714  831 ± 409  0.99  10.4 (9.0–11.8)  <0.0001 
 Unknown  13,480  11,537  938 ± 410  <0.0001  16.2 (15.5–16.8)  <0.0001 

aMean ± SD, adjusted for age, sex, and total energy intake, per 2,000 kcal for women and per 2,500 kcal for men.

bP values were corrected for multiple comparisons using the Bonferroni method.

A majority of lung cancer patients (78.1%) reported usual dietary calcium intakes from half to 1.5-fold of the RDA; 15.5% of patients consumed <0.5 RDA and 6.4% consumed >1.5 RDA. Low dietary calcium intake (<0.5 RDA) was associated with a small but significantly increased risk of death compared with the recommended calcium intake (800–1,200 mg/d; Table 2); the HRs (95% CIs) were 1.08 (1.03–1.14) in the model adjusted for age, sex, smoking status, pack-years, and total energy intake; and 1.07 (1.01–1.13) when further adjusted for known prognostic factors, including stage, histology, and grade. Supplemental calcium intake was not associated with lung cancer survival, although in the basic adjustment model, 200–500 mg/d supplemental calcium appeared to be associated with slightly reduced mortality (Table 2).

Table 2.

Pooled analyses of calcium intake and lung cancer survival (n = 23,882)

Hazard ratio (95% CI)
Calcium intakes Deaths/cases, n Model 1a Model 2b
Dietary calcium intake, mg/dc 
 <500 or <600  3,047/3,705  1.08 (1.03–1.14)  1.07 (1.01–1.13) 
 500–800 or 600–1,000  7,463/9,190  1.04 (1.00–1.08)  1.04 (1.00–1.09) 
 800–1,000 or 1,000–1,200  3,531/4,362  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  4,205/5,092  1.02 (0.97–1.07)  1.05 (1.01–1.10) 
 >1,500 or >1,800  1,292/1,533  1.03 (0.96–1.09)  1.04 (0.97–1.11) 
Dietary and supplemental calcium intake, mg/dc,d 
 <500 or <600  1,119/1,352  1.04 (0.96–1.11)  1.02 (0.95–1.10) 
 500–800 or 600–1000  4,333/5,212  1.01 (0.96–1.06)  0.99 (0.94–1.04) 
 800–1,000 or 1,000–1,200  2,695/3,216  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  4,364/5,290  0.97 (0.93–1.02)  1.01 (0.96–1.06) 
 >1,500 or >1,800  2,507/3,067  0.97 (0.91–1.02)  0.99 (0.94–1.05) 
Supplemental calcium intake, mg/dd 
 None  7,850/9,387  1.00 (ref)  1.00 (ref) 
 0–200  3,524/4,158  0.99 (0.95–1.04)  1.01 (0.97–1.06) 
 200–500  1,557/1,969  0.95 (0.90–1.00)  1.00 (0.94–1.05) 
 500–1,000  1,391/1,743  0.96 (0.91–1.02)  1.00 (0.94–1.06) 
 >1,000  696/880  0.97 (0.89–1.05)  1.01 (0.93–1.10) 
Hazard ratio (95% CI)
Calcium intakes Deaths/cases, n Model 1a Model 2b
Dietary calcium intake, mg/dc 
 <500 or <600  3,047/3,705  1.08 (1.03–1.14)  1.07 (1.01–1.13) 
 500–800 or 600–1,000  7,463/9,190  1.04 (1.00–1.08)  1.04 (1.00–1.09) 
 800–1,000 or 1,000–1,200  3,531/4,362  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  4,205/5,092  1.02 (0.97–1.07)  1.05 (1.01–1.10) 
 >1,500 or >1,800  1,292/1,533  1.03 (0.96–1.09)  1.04 (0.97–1.11) 
Dietary and supplemental calcium intake, mg/dc,d 
 <500 or <600  1,119/1,352  1.04 (0.96–1.11)  1.02 (0.95–1.10) 
 500–800 or 600–1000  4,333/5,212  1.01 (0.96–1.06)  0.99 (0.94–1.04) 
 800–1,000 or 1,000–1,200  2,695/3,216  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  4,364/5,290  0.97 (0.93–1.02)  1.01 (0.96–1.06) 
 >1,500 or >1,800  2,507/3,067  0.97 (0.91–1.02)  0.99 (0.94–1.05) 
Supplemental calcium intake, mg/dd 
 None  7,850/9,387  1.00 (ref)  1.00 (ref) 
 0–200  3,524/4,158  0.99 (0.95–1.04)  1.01 (0.97–1.06) 
 200–500  1,557/1,969  0.95 (0.90–1.00)  1.00 (0.94–1.05) 
 500–1,000  1,391/1,743  0.96 (0.91–1.02)  1.00 (0.94–1.06) 
 >1,000  696/880  0.97 (0.89–1.05)  1.01 (0.93–1.10) 

aCox model was stratified by cohort, year of lung cancer diagnosis, and time interval between dietary assessment and lung cancer diagnosis and adjusted for age at diagnosis, sex, smoking status, smoking pack-years, and total energy intake.

bAdditionally adjusted for race, education, alcohol consumption, history of diabetes, physical activity level, obesity status, hormone therapy in women, and the histological type, stage, and grade of lung cancer.

cFor men ≤70 years and women ≤50 years, the recommended dietary allowance (RDA) of calcium is 1,000 mg/d and the estimated average requirement (EAR) is 800 mg/d. For men >70 years and women >50 years, RDA is 1,200 mg/d and EAR is 1,000 mg/d. Calcium intakes were categorized into five groups: <0.5 RDA, 0.5 RDA to EAR, EAR to RDA, RDA to 1.5 RDA, and >1.5 RDA.

dSupplemental calcium intake data were available only in eight US cohorts, n = 18,137.

In stratified analysis, the association of low prediagnostic calcium intake with poor lung cancer survival was more evident in men than in women (Pinteraction = 0.01), and in early-stage than in distant-stage cases (Pinteraction = 0.006; Fig. 1). In particular, low calcium intake (<0.5 RDA vs. RDA) was associated with significantly increased mortality in male patients [HR (95% CI), 1.15 (1.06–1.25)], and in localized/regional stage patients [HR (95% CI), 1.15 (1.04–1.27)]. We did not observe significant interactions by other potential effect modifiers (Fig. 1). Kaplan–Meier survival curves by dietary calcium intakes, stratified by stage, histologic type, and grade, are shown in Supplementary Fig. S1.

Figure 1.

Risk of death by prediagnostic dietary calcium intake (low vs. recommended intake) in subgroups of the Calcium and Lung Cancer Pooling Project. Low intake was defined as calcium intake less than half of the recommended dietary allowance (RDA), which is less than 500 mg/d for men ≤70 years and women ≤50 years, or less than 600 mg/d for men >70 years and women >50 years. Recommended intake was defined as calcium intake between the estimated average requirement (EAR) and RDA, which is 800 to 1,000 mg/d for men ≤70 years and women ≤50 years, or 1,000 to 1,200 mg/d for men >70 years and women >50 years. The same stratified, multivariable-adjusted Cox model (Model 2) was used as shown in Table 2 footnote.

Figure 1.

Risk of death by prediagnostic dietary calcium intake (low vs. recommended intake) in subgroups of the Calcium and Lung Cancer Pooling Project. Low intake was defined as calcium intake less than half of the recommended dietary allowance (RDA), which is less than 500 mg/d for men ≤70 years and women ≤50 years, or less than 600 mg/d for men >70 years and women >50 years. Recommended intake was defined as calcium intake between the estimated average requirement (EAR) and RDA, which is 800 to 1,000 mg/d for men ≤70 years and women ≤50 years, or 1,000 to 1,200 mg/d for men >70 years and women >50 years. The same stratified, multivariable-adjusted Cox model (Model 2) was used as shown in Table 2 footnote.

Close modal

We thereafter focused our analyses among early-stage cases (Table 3, n = 8,013). Low dietary calcium intake (<0.5 RDA vs. RDA) was significantly associated with increased lung cancer mortality in early-stage patients, especially for men [HR (95% CI), 1.25 (1.08, 1.45)] and never smokers [HR (95% CI), 1.45 (1.01–2.08)]. Notably, we also observed that a very high calcium intake (>1.5 RDA vs. RDA) was associated with increased mortality in early-stage female patients [HR (95% CI), 1.33 (1.05–1.70)], although there were only 89 deaths and 134 female patients with a calcium intake this high. Among early-stage patients with low dietary calcium intake, supplemental calcium intake showed a possible trend of inverse association with mortality; compared with no or little supplemental calcium (<200 mg/d), HRs (95% CIs) were 0.90 (0.71–1.13) and 0.68 (0.44–1.07) for supplemental calcium of 200 to 1,000 and >1,000 mg/d, respectively. Meanwhile, among patients with no or little supplemental calcium intake, the HR (95% CI) for a low dietary calcium intake (<0.5 RDA vs. RDA) was 1.17 (1.02–1.35).

Table 3.

Dietary calcium intake and lung cancer survival in early-stage cases (n = 8,013)a

Hazard ratio (95% CI)
Dietary calcium intake, mg/d Deaths/cases, n Model 1 Model 2
All early-stage lung cancer cases 
 <500 or <600  928/1,342  1.12 (1.01–1.24)  1.15 (1.04–1.27) 
 500–800 or 600–1,000  2,083/3,065  1.06 (0.98–1.14)  1.06 (0.98–1.14) 
 800–1,000 or 1,000–1,200  990/1,442  1.00 (ref)  1.00 (ref) 
 1,000–1500 or 1,200–1,800  1,164/1,660  1.00 (0.92–1.09)  1.04 (0.96–1.14) 
 >1,500 or >1,800  371/504  0.99 (0.88–1.12)  1.07 (0.95–1.21) 
Female 
 <500 or <600  542/837  1.10 (0.95–1.28)  1.08 (0.92–1.25) 
 500–800 or 600–1,000  1,076/1,727  1.09 (0.96–1.24)  1.03 (0.90–1.17) 
 800–1,000 or 1,000–1,200  311/547  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  368/591  1.11 (0.95–1.30)  1.12 (0.96–1.31) 
 >1,500 or >1,800  89/134  1.25 (0.98–1.58)  1.33 (1.05–1.70) 
Male 
 <500 or <600  386/505  1.17 (1.01–1.36)  1.25 (1.08–1.45) 
 500–800 or 600–1,000  1,007/1,338  1.04 (0.94–1.15)  1.07 (0.97–1.18) 
 800–1,000 or 1,000–1,200  679/895  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  796/1,069  0.94 (0.85–1.05)  0.99 (0.90–1.10) 
 >1,500 or >1,800  282/370  0.93 (0.80–1.06)  0.99 (0.86–1.14) 
Whites 
 <500 or <600  507/691  1.12 (1.00–1.26)  1.16 (1.03–1.31) 
 500–800 or 600–1,000  1,673/2,377  1.05 (0.97–1.15)  1.06 (0.97–1.15) 
 800–1,000 or 1,000–1,200  873/1,254  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  1,103/1,539  1.02 (0.93–1.11)  1.05 (0.96–1.15) 
 >1,500 or >1,800  358/480  1.01 (0.89–1.15)  1.09 (0.96–1.23) 
Asians 
 <500 or <600  363/548  1.15 (0.97–1.35)  1.20 (1.01–1.43) 
 500–1,000 or 600–1,200  326/562  1.00 (ref)  1.00 (ref) 
 >1,000 or >1,200  33/76  0.73 (0.50–1.05)  0.76 (0.52–1.12) 
Blacks 
 <500 or <600  53/93  0.78 (0.55–1.09)  0.79 (0.55–1.14) 
 500–1,000 or 600–1,200  171/269  1.00 (ref)  1.00 (ref) 
 >1,000 or >1,200  25/47  0.60 (0.38–0.95)  0.77 (0.46–1.27) 
Never smokers 
 <500 or <600  143/260  1.32 (0.94–1.85)  1.45 (1.01–2.08) 
 500–800 or 600–1,000  204/410  1.06 (0.73–1.41)  1.12 (0.82–1.53) 
 800–1,000 or 1,000–1,200  71/146  1.00 (ref)  1.00 (ref) 
 1,000–1500 or 1,200–1,800  100/181  1.01 (0.73–1.40)  1.06 (0.76–1.49) 
 >1,500 or >1,800  20/41  0.84 (0.50–1.42)  0.86 (0.50–1.47) 
Former/current smokers 
 <500 or <600  785/1,082  1.09 (0.98–1.21)  1.12 (1.01–1.25) 
 500–800 or 600–1,000  1,879/2,655  1.06 (0.91–1.09)  1.05 (0.97–1.14) 
 800–1,000 or 1,000–1,200  919/1,296  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  1,064/1,479  1.00 (0.91–1.09)  1.04 (0.95–1.14) 
 >1,500 or >1,800  351/463  1.01 (0.89–1.15)  1.09 (0.96–1.23) 
Localized stage cases 
 <500 or <600  293/579  1.14 (0.96–1.37)  1.16 (0.97–1.40) 
 500–800 or 600–1,000  696/1,346  1.12 (0.97–1.29)  1.12 (0.97–1.29) 
 800–1,000 or 1,000–1,200  304/604  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  405/739  1.05 (0.91–1.23)  1.10 (0.95–1.28) 
 >1,500 or >1,800  125/221  0.93 (0.75–1.15)  0.97 (0.78–1.20) 
Regional stage cases 
 <500 or <600  635/763  1.07 (0.95–1.21)  1.10 (0.97–1.25) 
 500–800 or 600–1,000  1,387/1,719  1.01 (0.91–1.11)  1.02 (0.93–1.12) 
 800–1,000 or 1,000–1,200  686/838  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  759/921  1.00 (0.90–1.11)  1.01 (0.90–1.12) 
 >1,500 or >1,800  246/283  1.10 (0.95–1.27)  1.11 (0.96–1.29) 
Hazard ratio (95% CI)
Dietary calcium intake, mg/d Deaths/cases, n Model 1 Model 2
All early-stage lung cancer cases 
 <500 or <600  928/1,342  1.12 (1.01–1.24)  1.15 (1.04–1.27) 
 500–800 or 600–1,000  2,083/3,065  1.06 (0.98–1.14)  1.06 (0.98–1.14) 
 800–1,000 or 1,000–1,200  990/1,442  1.00 (ref)  1.00 (ref) 
 1,000–1500 or 1,200–1,800  1,164/1,660  1.00 (0.92–1.09)  1.04 (0.96–1.14) 
 >1,500 or >1,800  371/504  0.99 (0.88–1.12)  1.07 (0.95–1.21) 
Female 
 <500 or <600  542/837  1.10 (0.95–1.28)  1.08 (0.92–1.25) 
 500–800 or 600–1,000  1,076/1,727  1.09 (0.96–1.24)  1.03 (0.90–1.17) 
 800–1,000 or 1,000–1,200  311/547  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  368/591  1.11 (0.95–1.30)  1.12 (0.96–1.31) 
 >1,500 or >1,800  89/134  1.25 (0.98–1.58)  1.33 (1.05–1.70) 
Male 
 <500 or <600  386/505  1.17 (1.01–1.36)  1.25 (1.08–1.45) 
 500–800 or 600–1,000  1,007/1,338  1.04 (0.94–1.15)  1.07 (0.97–1.18) 
 800–1,000 or 1,000–1,200  679/895  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  796/1,069  0.94 (0.85–1.05)  0.99 (0.90–1.10) 
 >1,500 or >1,800  282/370  0.93 (0.80–1.06)  0.99 (0.86–1.14) 
Whites 
 <500 or <600  507/691  1.12 (1.00–1.26)  1.16 (1.03–1.31) 
 500–800 or 600–1,000  1,673/2,377  1.05 (0.97–1.15)  1.06 (0.97–1.15) 
 800–1,000 or 1,000–1,200  873/1,254  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  1,103/1,539  1.02 (0.93–1.11)  1.05 (0.96–1.15) 
 >1,500 or >1,800  358/480  1.01 (0.89–1.15)  1.09 (0.96–1.23) 
Asians 
 <500 or <600  363/548  1.15 (0.97–1.35)  1.20 (1.01–1.43) 
 500–1,000 or 600–1,200  326/562  1.00 (ref)  1.00 (ref) 
 >1,000 or >1,200  33/76  0.73 (0.50–1.05)  0.76 (0.52–1.12) 
Blacks 
 <500 or <600  53/93  0.78 (0.55–1.09)  0.79 (0.55–1.14) 
 500–1,000 or 600–1,200  171/269  1.00 (ref)  1.00 (ref) 
 >1,000 or >1,200  25/47  0.60 (0.38–0.95)  0.77 (0.46–1.27) 
Never smokers 
 <500 or <600  143/260  1.32 (0.94–1.85)  1.45 (1.01–2.08) 
 500–800 or 600–1,000  204/410  1.06 (0.73–1.41)  1.12 (0.82–1.53) 
 800–1,000 or 1,000–1,200  71/146  1.00 (ref)  1.00 (ref) 
 1,000–1500 or 1,200–1,800  100/181  1.01 (0.73–1.40)  1.06 (0.76–1.49) 
 >1,500 or >1,800  20/41  0.84 (0.50–1.42)  0.86 (0.50–1.47) 
Former/current smokers 
 <500 or <600  785/1,082  1.09 (0.98–1.21)  1.12 (1.01–1.25) 
 500–800 or 600–1,000  1,879/2,655  1.06 (0.91–1.09)  1.05 (0.97–1.14) 
 800–1,000 or 1,000–1,200  919/1,296  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  1,064/1,479  1.00 (0.91–1.09)  1.04 (0.95–1.14) 
 >1,500 or >1,800  351/463  1.01 (0.89–1.15)  1.09 (0.96–1.23) 
Localized stage cases 
 <500 or <600  293/579  1.14 (0.96–1.37)  1.16 (0.97–1.40) 
 500–800 or 600–1,000  696/1,346  1.12 (0.97–1.29)  1.12 (0.97–1.29) 
 800–1,000 or 1,000–1,200  304/604  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  405/739  1.05 (0.91–1.23)  1.10 (0.95–1.28) 
 >1,500 or >1,800  125/221  0.93 (0.75–1.15)  0.97 (0.78–1.20) 
Regional stage cases 
 <500 or <600  635/763  1.07 (0.95–1.21)  1.10 (0.97–1.25) 
 500–800 or 600–1,000  1,387/1,719  1.01 (0.91–1.11)  1.02 (0.93–1.12) 
 800–1,000 or 1,000–1,200  686/838  1.00 (ref)  1.00 (ref) 
 1,000–1,500 or 1,200–1,800  759/921  1.00 (0.90–1.11)  1.01 (0.90–1.12) 
 >1,500 or >1,800  246/283  1.10 (0.95–1.27)  1.11 (0.96–1.29) 

aThe same covariates and calcium cutoffs as shown in the footnote of Table 2. P for interaction was 0.02 for calcium intake levels with sex, 0.68 with race/ethnicity, 0.70 with smoking, and 0.47 with stage.

In cubic spline modeling, the lowest mortality among early-stage lung cancer patients was observed for dietary calcium intakes of 800 to 1200 mg/d (Fig. 2A, P = 0.03). Consistent with the above findings, low dietary calcium intake was associated with increased mortality, especially among early-stage male patients (Fig. 2B). Meanwhile, very high calcium intake may also be associated with increased mortality among early-stage female patients (Fig. 2C), although the confidence interval was very wide.

Figure 2.

Risk of death by prediagnostic dietary calcium intake in the Calcium and Lung Cancer Pooling Project (solid line, hazard ratio; dashed line, 95% confidence interval) among A, Early-stage lung cancer patients (P = 0.03); B, Early-stage male patients (P = 0.02); and C, early-stage female patients (P = 0.39). The same stratified, multivariable-adjusted Cox model (Model 2) was used as shown in Table 2 footnote.

Figure 2.

Risk of death by prediagnostic dietary calcium intake in the Calcium and Lung Cancer Pooling Project (solid line, hazard ratio; dashed line, 95% confidence interval) among A, Early-stage lung cancer patients (P = 0.03); B, Early-stage male patients (P = 0.02); and C, early-stage female patients (P = 0.39). The same stratified, multivariable-adjusted Cox model (Model 2) was used as shown in Table 2 footnote.

Close modal

Results were robust in sensitivity analyses and in meta-analysis. The HRs (95% CI) in early-stage cases for low dietary calcium intake were 1.14 (1.02–1.28) after excluding those diagnosed within two years after baseline (n = 6,362), 1.17 (1.05–1.30) after excluding those who died within three months after diagnosis (n = 7,246), 1.15 (1.02–1.28) for lung cancer–specific deaths (cases = 4,246), and 1.14 (1.02–1.28) in a fixed-effect meta-analysis (Pheterogeneity = 0.39; Supplementary Fig. S2).

In this large pooled analysis of 12 cohort studies, we observed that low prediagnostic dietary calcium intake (<500–600 mg/d) was associated with a slightly increased risk of death among lung cancer patients, after taking other prognostic factors into account. The lowest mortality was observed for dietary calcium intakes of 800 to 1,200 mg/d; further increase in calcium intake did not confer additional benefit. The association between low prediagnostic calcium intake and lung cancer survival was primarily confined to early-stage patients. No significant association was found for prediagnostic supplemental calcium intake with lung cancer survival.

Our study provides the first epidemiological evidence that long-term insufficient calcium intake may influence lung cancer prognosis, especially for early-stage patients, when cancer has not spread to other organs. Metastasis is the major reason for cancer-related deaths (50). For lung cancer, a frequent site of metastasis is the bone, occurring in nearly 40% of patients (51–53). Impaired bone metabolism and calcium homeostasis due to a long-term calcium insufficiency may facilitate bone metastasis and promote tumor growth in lung cancer. Calcium is an essential nutrient for bone remodeling (formation and resorption). A prolonged calcium deficiency leads to increased bone resorption and compromised bone health (11). When the circulating calcium level drops because of a low supply, multiple signaling pathways, hormones, and cytokines are affected. Directly, calcium is a second messenger and calcium signaling regulates cell differentiation, proliferation, and apoptosis (10, 54). Indirectly, to maintain a proper extracellular calcium level, the body upregulates secretions of PTH, 1,25-dihydroxyvitamin D, as well as several bone- or T cell–derived cytokines and growth factors, e.g., receptor activator of nuclear factor kappa-B ligand (RANKL), macrophage colony-stimulating factor, vascular endothelial growth factor, and interleukin-6 (55). Activation of these pathways enhances tumor growth, promotes angiogenesis, and accelerates metastasis (52, 55). During tumor progression, bone remodeling and calcium homeostasis are further disturbed. Tumor cells secrete factors that increase RANKL expression and bone resorption; in turn, growth factors released from the bone stimulate tumor growth and metastasis. This vicious cycle of bone destruction and tumor progression is well documented as an unfavorable prognostic factor of lung cancer (53, 56, 57). A few clinic-based studies have suggested that low blood calcium level and high bone resorption markers are associated with increased bone metastasis and decreased survival time among newly diagnosed lung cancer patients, after controlling for other clinicopathological factors (58–60). Therefore, bone-targeted therapies have been used to reduce bone metastases and prolong lung cancer survival (61, 62), and assessments of bone condition and bone metastasis have been recommended throughout lung cancer treatment (63, 64). Although limited by a lack of clinical information on patients' metastasis status or their blood calcium concentration or bone imaging data, our study is the first epidemiologic study to show that patients with habitually low calcium intakes were at a high risk of death. Our findings suggest that assessments of bone health and calcium homeostasis may benefit these patients, especially those diagnosed at early stages, by allowing for a proper intervention.

A U-shaped relationship was found in recent meta-analyses of calcium intake with mortality from all causes, cardiovascular disease, and cancer (65, 66). Our results also suggest a possible U-shaped association between dietary calcium and lung cancer survival with an optimal intake around 1,000 mg/d; intakes of <500 mg/d (especially among early-stage men) or >1,800 mg/d (especially among early-stage women) were both associated with increased mortality. However, the number of patients with excessive dietary calcium intake was too small to draw conclusions from this observation. In addition, the increased risk among very high calcium consumers may indirectly reflect a history of calcium deficiency and poor bone health, leading to a subsequent increase in calcium consumption, particularly for women who are more likely to be affected by and diagnosed with osteoporosis. Although the sex-differential associations we observed could be due to chance, they warrant further investigation. Possible explanations include gender differences in calcium intake level, lung cancer histology, estrogen exposure, and lifestyle habits.

Our study did not find a significant association between supplemental calcium intake and lung cancer survival among the 18,137 incident cases from eight US cohorts that collected this information. The null association may be due to the fact that fewer individuals would have an insufficient calcium intake when taking calcium supplements. If only a long-term low calcium intake was associated with cancer prognosis, a null association for supplemental calcium would be expected. We did observe a possible trend of beneficial association for supplemental calcium among early-stage patients with low dietary calcium intakes (see Results section). The null finding could also be due to a suboptimal measurement of usual supplement intake. Nevertheless, this finding is in line with results from randomized, controlled trials which found no significant effects of calcium supplements on cancer incidence or mortality (67–69). Taken together, our results support the hypothesis that an optimal calcium intake from foods may favorably impact cancer survival; however, evidence is lacking for a recommendation to use calcium supplements to increase intake, especially among those already consuming a sufficient amount of dietary calcium.

Our study has several limitations. First, dietary intakes were measured via FFQs and food composition tables, both of which have nonnegligible measurement errors. Although FFQs used in participating cohorts have each been validated and shown reasonably good validities (37–45, 47, 48), measurement errors cannot be precluded. Nondifferential misclassifications of calcium intakes may bias our risk ratio estimates toward the null. Second, information on post-diagnostic calcium intake, comorbidity status at the time of diagnosis, and cancer treatment was unavailable, and tumor stage and grade data were missing in a sizable fraction of patients. Cancer treatment and comorbidities may affect calcium homeostasis, which may exacerbate the hazardous effect of a preexisting low calcium status. Lack of treatment and comorbidity information prevented us from investigating this hypothesis. Most of our study patients were diagnosed before 2010 when targeted treatments were less common and lung cancer treatments were largely based on stage, histology, and patient sociodemographics. We adjusted for all these factors, as well as baseline diabetes and obesity status, and stratified by cohort (region) and year of diagnosis. We also conducted a number of subgroup analyses and did not find significant effect modifications except by stage. Moreover, patient tumor characteristics (including the unknown category) were not associated with prediagnostic calcium intake, suggesting that these clinical factors were unlikely to substantially confound the association. Third, despite the large sample size, statistical power remained limited in certain subgroup analyses, such as the interaction analysis by race/ethnicity and the analysis among never smokers. For example, we found a seemingly opposite association in blacks, but the small number of black patients and the insignificant interaction with race/ethnicity prevented us from drawing any conclusions. Finally, we cannot separate the effects of calcium from related nutrients, including vitamin D, magnesium, phosphorus, and other nutrients in calcium-rich foods, nor could we rule out residual confounding from unknown confounders and imperfectly measured covariates, which may be particularly challenging in pooling projects using harmonized data from multiple studies.

Strengths of our study include its prospective design, large sample size, and pooled data analysis. The prospective study design minimized reverse causality and biases of recall and selection. Pooled individual-level data from 12 cohort studies in three continents enabled us to evaluate the associations among populations with a broad range of exposures and by clinical characteristics, and to examine calcium intake via multiple approaches, i.e., using project-wide cutoff points, as continuous variables, and in a series of sensitivity analyses. Results for low dietary calcium intake and poorer survival were largely consistent when using different approaches.

In summary, in this large pooled analysis, we found that low prediagnostic dietary calcium intake (<500–600 mg/d) was associated with a small increased risk of death among localized and regional stage lung cancer patients. Further studies with biomarker data and more detailed clinical data are needed to confirm or refute our findings. Meanwhile, studies are needed to explore biological mechanisms linking calcium nutrition, calcium homeostasis, and bone remodeling with lung cancer progression, as well as to investigate modifiable nutrition and lifestyle factors to improve survival for lung cancer.

No potential conflicts of interest were disclosed.

Conception and design: D. Yu, Y. Takata, E. Giovannucci, Y. Park, A. Langhammer, E. Weiderpass, K. Hveem, X.-O. Shu

Development of methodology: D. Yu, A. Langhammer, E. Weiderpass, S. Krokstad, X.-O. Shu

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): D. Yu, S.A. Smith-Warner, W. Blot, N. Sawada, E. White, N. Freedman, K. Robien, E. Giovannucci, X. Zhang, Y. Park, Y.-T. Gao, R.T. Chlebowski, A. Langhammer, G. Yang, G. Severi, J. Manjer, K.-T. Khaw, E. Weiderpass, S. Krokstad, K. Hveem, R. Sinha, R. Ziegler, S. Tsugane, Y.-B. Xiang, M. Johansson, W. Zheng, X.-O. Shu

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): D. Yu, Y. Takata, S.A. Smith-Warner, N. Freedman, X. Zhang, Y. Park, R.T. Chlebowski, E. Weiderpass, Y.-B. Xiang

Writing, review, and/or revision of the manuscript: D. Yu, Y. Takata, S.A. Smith-Warner, W. Blot, E. White, N. Freedman, K. Robien, E. Giovannucci, X. Zhang, Y. Park, Y.-T. Gao, R.T. Chlebowski, A. Langhammer, G. Yang, G. Severi, J. Manjer, K.-T. Khaw, E. Weiderpass, L.M. Liao, N. Caporaso, S. Krokstad, K. Hveem, R. Sinha, R. Ziegler, S. Tsugane, Y.-B. Xiang, M. Johansson, W. Zheng, X.-O. Shu

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): N. Sawada, N. Freedman, K. Robien, Y.-T. Gao, J. Manjer, K.-T. Khaw, E. Weiderpass, L.M. Liao, S. Tsugane, Y.-B. Xiang, M. Johansson

Study supervision: Y.-T. Gao, E. Weiderpass, X.-O. Shu

We thank the staff and investigators of all participating cohorts for their dedicated efforts. We thank Ms. Nancy Kennedy for her assistance in editing and preparing the manuscript.

This work was supported by a grant from the NCI at the NIH to Drs. X.-O. Shu and Y. Takata (R03 CA183021). The NCI had no role in the design and conduct of the study; collection, management, analysis, or interpretation of the data; preparation, review, or approval of the manuscript; or decision to submit the manuscript for publication.

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

1.
American Cancer Society
.
Global cancer facts and figures. 3rd ed
.
Atlanta
:
American Cancer Society
; 
2015
.
2.
Howlader
N
,
Noone
A
,
Krapcho
M
,
Garshell
J
,
Miller
D
,
Altekruse
S
, et al
SEER Cancer Statistics Review, 1975-2012
.
[Internet]
.
Bethesda, MD
:
National Cancer Institute
; 
2015
.
Available from
: http://seer.cancer.gov/csr/1975_2012/
3.
Brenner
H
,
Francisci
S
,
de Angelis
R
,
Marcos-Gragera
R
,
Verdecchia
A
,
Gatta
G
, et al
Long-term survival expectations of cancer patients in Europe in 2000–2002
.
Eur J Cancer
2009
;
45
:
1028
41
.
4.
Zeng
H
,
Zheng
R
,
Guo
Y
,
Zhang
S
,
Zou
X
,
Wang
N
, et al
Cancer survival in China, 2003–2005: a population-based study
.
Int J Cancer
2015
;
136
:
1921
30
.
5.
Sculier
J-P
,
Chansky
K
,
Crowley
JJ
,
Van Meerbeeck
J
,
Goldstraw
P
,
International Staging Committee and Participating Institutions
. 
The impact of additional prognostic factors on survival and their relationship with the anatomical extent of disease expressed by the 6th Edition of the TNM Classification of Malignant Tumors and the proposals for the 7th Edition
.
J Thorac Oncol Off Publ Int Assoc Study Lung Cancer
2008
;
3
:
457
66
.
6.
Zeng
C
,
Wen
W
,
Morgans
AK
,
Pao
W
,
Shu
X-O
,
Zheng
W
. 
Disparities by race, age, and sex in the improvement of survival for major cancers: results from the National Cancer Institute Surveillance, Epidemiology, and End Results (SEER) Program in the United States, 1990 to 2010
.
JAMA Oncol
2015
;
1
:
88
96
.
7.
Yang
G
,
Shu
X-O
,
Li
H-L
,
Chow
W-H
,
Wen
W
,
Xiang
Y-B
, et al
Prediagnosis soy food consumption and lung cancer survival in women
.
J Clin Oncol
2013
;
31
:
1548
53
.
8.
Wu
Q-J
,
Yang
G
,
Zheng
W
,
Li
H-L
,
Gao
J
,
Wang
J
, et al
Pre-diagnostic cruciferous vegetables intake and lung cancer survival among Chinese women
.
Sci Rep
2015
;
5
:
10306
.
9.
Hasegawa
Y
,
Ando
M
,
Maemondo
M
,
Yamamoto
S
,
Isa
S-I
,
Saka
H
, et al
The role of smoking status on the progression-free survival of non-small cell lung cancer patients harboring activating epidermal growth factor receptor (EGFR) mutations receiving first-line EGFR tyrosine kinase inhibitor versus platinum doublet chemotherapy: a meta-analysis of prospective randomized trials
.
The Oncologist
2015
;
20
:
307
15
.
10.
Peterlik
M
,
Grant
WB
,
Cross
HS
. 
Calcium, vitamin D and cancer
.
Anticancer Res
2009
;
29
:
3687
98
.
11.
Institute of Medicine (US) Committee to Review Dietary Reference Intakes for Vitamin D and Calcium
.
Dietary Reference Intakes for Calcium and Vitamin D [Internet]
.
Ross
AC
,
Taylor
CL
,
Yaktine
AL
,
Del Valle
HB
,
editors
.
Washington (DC)
:
National Academies Press (US)
; 
2011
[cited 2015 Nov 24]. Available from
: http://www.ncbi.nlm.nih.gov/books/NBK56070/.
12.
Peterlik
M
,
Kállay
E
,
Cross
HS
. 
Calcium nutrition and extracellular calcium sensing: relevance for the pathogenesis of osteoporosis, cancer and cardiovascular diseases
.
Nutrients
2013
;
5
:
302
27
.
13.
Park
Y
,
Leitzmann
MF
,
Subar
AF
,
Hollenbeck
A
,
Schatzkin
A
. 
Dairy food, calcium, and risk of cancer in the NIH-AARP Diet and Health Study
.
Arch Intern Med
2009
;
169
:
391
401
.
14.
Cho
E
,
Smith-Warner
SA
,
Spiegelman
D
,
Beeson
WL
,
van den Brandt
PA
,
Colditz
GA
, et al
Dairy foods, calcium, and colorectal cancer: a pooled analysis of 10 cohort studies
.
J Natl Cancer Inst
2004
;
96
:
1015
22
.
15.
Huncharek
M
,
Muscat
J
,
Kupelnick
B
. 
Colorectal cancer risk and dietary intake of calcium, vitamin D, and dairy products: a meta-analysis of 26,335 cases from 60 observational studies
.
Nutr Cancer
2009
;
61
:
47
69
.
16.
Lin
J
,
Manson
JE
,
Lee
I-M
,
Cook
NR
,
Buring
JE
,
Zhang
SM
. 
Intakes of calcium and vitamin D and breast cancer risk in women
.
Arch Intern Med
2007
;
167
:
1050
9
.
17.
Chen
P
,
Hu
P
,
Xie
D
,
Qin
Y
,
Wang
F
,
Wang
H
. 
Meta-analysis of vitamin D, calcium and the prevention of breast cancer
.
Breast Cancer Res Treat
2010
;
121
:
469
77
.
18.
Wilson
KM
,
Shui
IM
,
Mucci
LA
,
Giovannucci
E
. 
Calcium and phosphorus intake and prostate cancer risk: a 24-y follow-up study
.
Am J Clin Nutr
2015
;
101
:
173
83
.
19.
Aune
D
,
Navarro Rosenblatt
DA
,
Chan
DSM
,
Vieira
AR
,
Vieira
R
,
Greenwood
DC
, et al
Dairy products, calcium, and prostate cancer risk: a systematic review and meta-analysis of cohort studies
.
Am J Clin Nutr
2015
;
101
:
87
117
.
20.
Mahabir
S
,
Forman
MR
,
Dong
YQ
,
Park
Y
,
Hollenbeck
A
,
Schatzkin
A
. 
Mineral intake and lung cancer risk in the NIH-American Association of Retired Persons Diet and Health study
.
Cancer Epidemiol Biomark Prev
2010
;
19
:
1976
83
.
21.
Li
K
,
Kaaks
R
,
Linseisen
J
,
Rohrmann
S
. 
Dietary calcium and magnesium intake in relation to cancer incidence and mortality in a German prospective cohort (EPIC-Heidelberg)
.
Cancer Causes Control
2011
;
22
:
1375
82
.
22.
Takata
Y
,
Shu
X-O
,
Yang
G
,
Li
H
,
Dai
Q
,
Gao
J
, et al
Calcium intake and lung cancer risk among female nonsmokers: a report from the Shanghai Women's Health Study
.
Cancer Epidemiol Biomark Prev
2013
;
22
:
50
7
.
23.
Dik
VK
,
Murphy
N
,
Siersema
PD
,
Fedirko
V
,
Jenab
M
,
Kong
SY
, et al
Prediagnostic intake of dairy products and dietary calcium and colorectal cancer survival–results from the EPIC cohort study
.
Cancer Epidemiol Biomark Prev
2014
;
23
:
1813
23
.
24.
Yang
B
,
McCullough
ML
,
Gapstur
SM
,
Jacobs
EJ
,
Bostick
RM
,
Fedirko
V
, et al
Calcium, vitamin D, dairy products, and mortality among colorectal cancer survivors: the Cancer Prevention Study-II Nutrition Cohort
.
J Clin Oncol
2014
;
32
:
2335
43
.
25.
Schatzkin
A
,
Subar
AF
,
Thompson
FE
,
Harlan
LC
,
Tangrea
J
,
Hollenbeck
AR
, et al
Design and serendipity in establishing a large cohort with wide dietary intake distributions: the National Institutes of Health-American Association of Retired Persons Diet and Health Study
.
Am J Epidemiol
2001
;
154
:
1119
25
.
26.
Michaud
DS
,
Liu
Y
,
Meyer
M
,
Giovannucci
E
,
Joshipura
K
. 
Periodontal disease, tooth loss, and cancer risk in male health professionals: a prospective cohort study
.
Lancet Oncol
2008
;
9
:
550
8
.
27.
Baik
CS
,
Strauss
GM
,
Speizer
FE
,
Feskanich
D
. 
Reproductive factors, hormone use, and risk for lung cancer in postmenopausal women, the Nurses' Health Study
.
Cancer Epidemiol Biomark Prev
2010
;
19
:
2525
33
.
28.
Sinner
P
,
Folsom
AR
,
Harnack
L
,
Eberly
LE
,
Schmitz
KH
. 
The association of physical activity with lung cancer incidence in a cohort of older women: the Iowa Women's Health Study
.
Cancer Epidemiol Biomark Prev
2006
;
15
:
2359
63
.
29.
Zhu
CS
,
Pinsky
PF
,
Kramer
BS
,
Prorok
PC
,
Purdue
MP
,
Berg
CD
, et al
The prostate, lung, colorectal, and ovarian cancer screening trial and its associated research resource
.
J Natl Cancer Inst
2013
;
105
:
1684
93
.
30.
Signorello
LB
,
Hargreaves
MK
,
Blot
WJ
. 
The Southern Community Cohort Study: investigating health disparities
.
J Health Care Poor Underserved
2010
;
21
:
26
37
.
31.
White
E
,
Patterson
RE
,
Kristal
AR
,
Thornquist
M
,
King
I
,
Shattuck
AL
, et al
VITamins And Lifestyle cohort study: study design and characteristics of supplement users
.
Am J Epidemiol
2004
;
159
:
83
93
.
32.
Langer
RD
,
White
E
,
Lewis
CE
,
Kotchen
JM
,
Hendrix
SL
,
Trevisan
M
. 
The women's health initiative observational study: baseline characteristics of participants and reliability of baseline measures
.
Ann Epidemiol
2003
;
13
:
S107
21
.
33.
Riboli
E
,
Hunt
KJ
,
Slimani
N
,
Ferrari
P
,
Norat
T
,
Fahey
M
, et al
European Prospective Investigation into Cancer and Nutrition (EPIC): study populations and data collection
.
Public Health Nutr
2002
;
5
:
1113
24
.
34.
Tsugane
S
,
Sawada
N
. 
The JPHC study: design and some findings on the typical Japanese diet
.
Jpn J Clin Oncol
2014
;
44
:
777
82
.
35.
Shu
X-O
,
Li
H
,
Yang
G
,
Gao
J
,
Cai
H
,
Takata
Y
, et al
Cohort Profile: The Shanghai Men's Health Study
.
Int J Epidemiol
2015
;
dyv013
.
36.
Zheng
W
,
Chow
W-H
,
Yang
G
,
Jin
F
,
Rothman
N
,
Blair
A
, et al
The Shanghai Women's Health Study: rationale, study design, and baseline characteristics
.
Am J Epidemiol
2005
;
162
:
1123
31
.
37.
Thompson
FE
,
Kipnis
V
,
Midthune
D
,
Freedman
LS
,
Carroll
RJ
,
Subar
AF
, et al
Performance of a food-frequency questionnaire in the US NIH-AARP (National Institutes of Health-American Association of Retired Persons) Diet and Health Study
.
Public Health Nutr
2008
;
11
:
183
95
.
38.
Willett
WC
,
Sampson
L
,
Stampfer
MJ
,
Rosner
B
,
Bain
C
,
Witschi
J
, et al
Reproducibility and validity of a semiquantitative food frequency questionnaire
.
Am J Epidemiol
1985
;
122
:
51
65
.
39.
Rimm
EB
,
Giovannucci
EL
,
Stampfer
MJ
,
Colditz
GA
,
Litin
LB
,
Willett
WC
. 
Reproducibility and validity of an expanded self-administered semiquantitative food frequency questionnaire among male health professionals
.
Am J Epidemiol
1992
;
135
:1114–
1126
1136
.
40.
Munger
RG
,
Folsom
AR
,
Kushi
LH
,
Kaye
SA
,
Sellers
TA
. 
Dietary assessment of older Iowa women with a food frequency questionnaire: nutrient intake, reproducibility, and comparison with 24-hour dietary recall interviews
.
Am J Epidemiol
1992
;
136
:
192
200
.
41.
Subar
AF
,
Thompson
FE
,
Kipnis
V
,
Midthune
D
,
Hurwitz
P
,
McNutt
S
, et al
Comparative Validation of the Block, Willett, and National Cancer Institute Food Frequency Questionnaires The Eating at America's Table Study
.
Am J Epidemiol
2001
;
154
:
1089
99
.
42.
Signorello
LB
,
Munro
HM
,
Buchowski
MS
,
Schlundt
DG
,
Cohen
SS
,
Hargreaves
MK
, et al
Estimating nutrient intake from a food frequency questionnaire: incorporating the elements of race and geographic region
.
Am J Epidemiol
2009
;
170
:
104
11
.
43.
Patterson
RE
,
Kristal
AR
,
Tinker
LF
,
Carter
RA
,
Bolton
MP
,
Agurs-Collins
T
. 
Measurement characteristics of the Women's Health Initiative food frequency questionnaire
.
Ann Epidemiol
1999
;
9
:
178
87
.
44.
Slimani
N
,
Ferrari
P
,
Ocké
M
,
Welch
A
,
Boeing
H
,
Liere
M
, et al
Standardization of the 24-hour diet recall calibration method used in the european prospective investigation into cancer and nutrition (EPIC): general concepts and preliminary results
.
Eur J Clin Nutr
2000
;
54
:
900
17
.
45.
Ishihara
J
,
Sobue
T
,
Yamamoto
S
,
Yoshimi
I
,
Sasaki
S
,
Kobayashi
M
, et al
Validity and reproducibility of a self-administered food frequency questionnaire in the JPHC Study Cohort II: study design, participant profile and results in comparison with Cohort I
.
J Epidemiol Jpn Epidemiol Assoc
2003
;
13
:
S134
47
.
46.
Tsubono
Y
,
Kobayashi
M
,
Sasaki
S
,
Tsugane
S
,
JPHC
. 
Validity and reproducibility of a self-administered food frequency questionnaire used in the baseline survey of the JPHC Study Cohort I
.
J Epidemiol Jpn Epidemiol Assoc
2003
;
13
:
S125
33
.
47.
Villegas
R
,
Yang
G
,
Liu
D
,
Xiang
Y-B
,
Cai
H
,
Zheng
W
, et al
Validity and reproducibility of the food-frequency questionnaire used in the Shanghai men's health study
.
Br J Nutr
2007
;
97
:
993
1000
.
48.
Shu
XO
,
Yang
G
,
Jin
F
,
Liu
D
,
Kushi
L
,
Wen
W
, et al
Validity and reproducibility of the food frequency questionnaire used in the Shanghai Women's Health Study
.
Eur J Clin Nutr
2004
;
58
:
17
23
.
49.
Willett
WC
,
Howe
GR
,
Kushi
LH
. 
Adjustment for total energy intake in epidemiologic studies
.
Am J Clin Nutr
1997
;
65
:
1220S
1228S
.
50.
Mehlen
P
,
Puisieux
A
. 
Metastasis: a question of life or death
.
Nat Rev Cancer
2006
;
6
:
449
58
.
51.
Coleman
RE
. 
Clinical features of metastatic bone disease and risk of skeletal morbidity
.
Clin Cancer Res
2006
;
12
:
6243s
6249s
.
52.
Brodowicz
T
,
O'Byrne
K
,
Manegold
C
. 
Bone matters in lung cancer
.
Ann Oncol
2012
;
23
:
2215
22
.
53.
Riihimäki
M
,
Hemminki
A
,
Fallah
M
,
Thomsen
H
,
Sundquist
K
,
Sundquist
J
, et al
Metastatic sites and survival in lung cancer
.
Lung Cancer Amst Neth
2014
;
86
:
78
84
.
54.
Yang
H
,
Zhang
Q
,
He
J
,
Lu
W
. 
Regulation of calcium signaling in lung cancer
.
J Thorac Dis
2010
;
2
:
52
6
.
55.
Roodman
GD
. 
Mechanisms of bone metastasis
.
N Engl J Med
2004
;
350
:
1655
64
.
56.
Cetin
K
,
Christiansen
CF
,
Jacobsen
JB
,
Nørgaard
M
,
Sørensen
HT
. 
Bone metastasis, skeletal-related events, and mortality in lung cancer patients: a Danish population-based cohort study
.
Lung Cancer
2014
;
86
:
247
54
.
57.
Mountzios
G
,
Ramfidis
V
,
Terpos
E
,
Syrigos
KN
. 
Prognostic significance of bone markers in patients with lung cancer metastatic to the skeleton: a review of published data
.
Clin Lung Cancer
2011
;
12
:
341
9
.
58.
Shen
H
,
Li
Y
,
Liao
Y
,
Zhang
T
,
Liu
Q
,
Du
J
. 
Lower blood calcium associates with unfavorable prognosis and predicts for bone metastasis in NSCLC
.
PLoS One
2012
;
7
:
e34264
.
59.
Tamiya
M
,
Kobayashi
M
,
Morimura
O
,
Yasue
T
,
Nakasuji
T
,
Satomu
M
, et al
Clinical significance of the serum crosslinked N-telopeptide of type I collagen as a prognostic marker for non-small-cell lung cancer
.
Clin Lung Cancer
2013
;
14
:
50
4
.
60.
Wang
Z
,
Lu
Y
,
Qiao
D
,
Wen
X
,
Zhao
H
,
Yao
Y
. 
Diagnostic and prognostic validity of serum bone turnover markers in bone metastatic non-small cell lung cancer patients
.
J Bone Oncol
2015
;
4
:
85
91
.
61.
Lopez-Olivo
MA
,
Shah
NA
,
Pratt
G
,
Risser
JM
,
Symanski
E
,
Suarez-Almazor
ME
. 
Bisphosphonates in the treatment of patients with lung cancer and metastatic bone disease: a systematic review and meta-analysis
.
Support Care Cancer
2012
;
20
:
2985
98
.
62.
De Castro
J
,
García
R
,
Garrido
P
,
Isla
D
,
Massuti
B
,
Blanca
B
, et al
Therapeutic potential of denosumab in patients with lung cancer: beyond prevention of skeletal complications
.
Clin Lung Cancer
2015
;
16
:
431
46
.
63.
De Marinis
F
,
Eberhardt
W
,
Harper
PG
,
Sureda
BM
,
Nackaerts
K
,
Soerensen
JB
, et al
Bisphosphonate use in patients with lung cancer and bone metastases: recommendations of a European expert panel
.
J Thorac Oncol
2009
;
4
:
1280
8
.
64.
Langer
C
,
Hirsh
V
. 
Skeletal morbidity in lung cancer patients with bone metastases: demonstrating the need for early diagnosis and treatment with bisphosphonates
.
Lung Cancer
2010
;
67
:
4
11
.
65.
Wang
X
,
Chen
H
,
Ouyang
Y
,
Liu
J
,
Zhao
G
,
Bao
W
, et al
Dietary calcium intake and mortality risk from cardiovascular disease and all causes: a meta-analysis of prospective cohort studies
.
BMC Med
2014
;
12
:
158
.
66.
Asemi
Z
,
Saneei
P
,
Sabihi
S-S
,
Feizi
A
,
Esmaillzadeh
A
. 
Total, dietary, and supplemental calcium intake and mortality from all-causes, cardiovascular disease, and cancer: a meta-analysis of observational studies
.
Nutr Metab Cardiovasc Dis
2015
;
25
:
623
34
.
67.
Brunner
RL
,
Wactawski-Wende
J
,
Caan
BJ
,
Cochrane
BB
,
Chlebowski
RT
,
Gass
MLS
, et al
The effect of calcium plus vitamin D on risk for invasive cancer: results of the Women's Health Initiative (WHI) calcium plus vitamin D randomized clinical trial
.
Nutr Cancer
2011
;
63
:
827
41
.
68.
Avenell
A
,
MacLennan
GS
,
Jenkinson
DJ
,
McPherson
GC
,
McDonald
AM
,
Pant
PR
, et al
Long-term follow-up for mortality and cancer in a randomized placebo-controlled trial of vitamin D(3) and/or calcium (RECORD trial)
.
J Clin Endocrinol Metab
2012
;
97
:
614
22
.
69.
Bolland
MJ
,
Grey
A
,
Gamble
GD
,
Reid
IR
. 
The effect of vitamin D supplementation on skeletal, vascular, or cancer outcomes: a trial sequential meta-analysis
.
Lancet Diabetes Endocrinol
2014
;
2
:
307
20
.