Background:

Insulin-like growth factor-1 (IGF-1) has been implicated in several malignancies, but few studies have examined multiple cancers simultaneously. We sought to conduct systematic assessments of the association between IGF-1 and cancer risk.

Methods:

We conducted a prospective analysis between IGF-1 and incident total and 19 site-specific cancers among 412,645 individuals enrolled in the UK Biobank with follow-up to 2016. IGF-1 was measured using blood samples provided at the baseline examination. HR and 95% confidence interval (CI) were calculated with multivariable-adjusted Cox models with IGF-1 modeled both in sex-specific quintiles and continuously.

Results:

Participants were followed for a median of 7.2 years. We observed positive associations between circulating IGF-1 and overall cancer risk for both men (HR = 1.03 per 5-nmol/L increment in IGF-1; 95% CI, 1.01–1.06) and women (HR = 1.03; 95% CI, 1.01–1.06). For specific sites, we observed positive associations for breast (HR = 1.10; 95% CI, 1.07–1.14), prostate (1.09; 95% CI, 1.05–1.12), colorectum (1.07; 95% CI, 1.02–1.11), melanoma (1.08; 95% CI, 1.01–1.15), kidney (1.10; 95% CI, 1.00–1.20), and thyroid (1.22; 95% CI, 1.05–1.42) and inverse associations for lung (0.91; 95% CI, 0.86–0.96), ovaries (0.86; 95% CI, 0.77–0.95), head and neck (0.90; 95% CI, 0.82–0.99), and liver (0.32; 95% CI, 0.26–0.38). The inverse association between IGF-1 and lung cancer was observed only in ever-smokers (HRever-smoker = 0.88 vs. HRnever-smoker = 1.14; Pinteraction = 0.0005). Analyses comparing extreme quintiles were consistent.

Conclusions:

IGF-1 is modestly associated with increased risk of total cancer in both men and women but demonstrated divergent associations for site-specific cancers.

Impact:

Our study suggests that IGF-1 could serve as a target for cancer prevention or treatment.

The insulin-like growth factor (IGF) axis plays a crucial role in human growth and development (1). IGF-1 levels signal important early life exposures and are associated with childhood adiposity and height velocity. Perhaps due to its influence on growth, mitosis, and apoptosis, higher circulating IGF-1 has been implicated in multiple malignancies in observational epidemiologic studies, including for breast (2–4), prostate (5, 6), colorectal (7), lung (8), ovarian (9), and central nervous system cancers (glioma; ref. 10). In addition, recent epidemiologic studies suggest that metformin usage may reduce breast cancer incidence and improve overall survival, which may be mediated by metformin's inhibition of the IGF-1 pathway (11, 12). Similarly, a phase II randomized controlled trial (RCT) demonstrated improved survival in patients with lung cancer who received metformin in addition to standard therapy (13). These findings raise interesting questions for the IGF-1 pathway as a biomarker for incident cancer and as a potential target for cancer prevention or therapeutics. However, most existing observational studies were conducted around the 2000s, were relatively limited by sample size or study design (retrospective case–control studies), were inconsistent in their findings, did not comprehensively assess the association of IGF-1 with cancers at uncommon cancer sites, nor examined demographic or clinical characteristics that may modify the associations between IGF-1 and individual cancers.

In turn, we sought to conduct a comprehensive analysis of circulating IGF-1 and incident total and site-specific cancers in the UK Biobank cohort, which is comprised of approximately 500,000 participants. We hypothesized that IGF-1 is positively associated with total and most site-specific cancers.

Study population

The UK Biobank cohort recruited 502,656 adult participants, ages 40 to 69, between 2007 and 2010 (14), from approximately 9.2 million people who were invited to participate (response rate: 5.7%). All participants were registered with the UK National Health Service (NHS) and lived within approximately 25 miles of one of the 22 study centers. At the baseline examination, participants completed questionnaires regarding their demographic, lifestyle, disease histories, and medication use and provided biospecimens. Blood samples were collected using standard vacutainers, transported overnight to a central laboratory, where the blood was centrifuged and aliquots of specific fractions were stored at −80°C, prior to biomarker measurements and genotyping (15).

We excluded participants if they reported a history of the cancer being assessed (for the analysis on total cancers, participants with prior diagnosis of any cancer was excluded), prevalent type 2 diabetes or unknown diabetes status at recruitment (due to the potential for hypoglycemic agents to affect IGF-1 levels; N = 25,345; ref. 16), current oral contraceptive or menopausal hormone use at the baseline exam [oral estrogens can affect circulating levels of multiple hormones, including IGF system markers (16), through a first pass effect on the liver; N = 24,448], missing information on genetic ancestry (N = 4,249), discordant self-reported and genetic sex (N = 355), and participants without a baseline IGF-1 measurement (N = 35,464). In addition, 21 participants withdrew consent prior to the submission of this manuscript and were removed from the analysis. Hence, our final analytical sample consisted of 412,645 participants, prior to the exclusion of participants with prevalent cancer.

Biomarker measurements

Circulating IGF-1 were measured in the serum of stored blood samples from 502,527 participants using a chemiluminescent immunoassay with the Liaison XL platform (DiaSorin Ltd), as part of the UK Biobank Biomarker Project (17). Quality control procedures and results were published previously (17). Briefly, average within-laboratory (total) coefficient of variation (CV) for low, medium, and high internal quality control level samples for each biomarker ranged from 1.7% to 15.3% (for IGF-1, the CVs ranged from 5.3% to 6.2%; ref. 17). IGF-1 was measured at both the initial and a follow-up visit (median ∼4 years apart, using blood samples collected in 2012–2013) in 16,357 participants. These participants constitute a subset of the original cohort, who live in the area surrounding the UK Biobank's coordinating center in Stockport, and voluntarily chose to undergo a repeat physical examination, update their answers to the original questionnaires, and provide biospecimens.

Testosterone and sex hormone binding globulin (SHBG) were determined by a chemiluminescent immunoassay using the Beckman Coulter DXI 800. High-sensitivity C-reactive protein (hsCRP) levels were measured with the immunoturbidimetric method (Beckman Coulter DXI 800). Glycated hemoglobin (HbA1c) levels were determined using the HPLC Variant II Turbo 2.0 system (Bio-Rad). Alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyltransferase (GGT) were assayed using enzymatic rate measurements (Beckman Coulter AU5800). Total bilirubin was measured with colorimetric analysis (Beckman Coulter AU5800).

Disease ascertainment

Linkages to NHS medical records with the International Classification of Diseases (ICD-10) diagnosis codes were used to ascertain a cancer diagnosis (18). Since the last data update, cancer diagnoses follow-up is available through March 31, 2016 for participants in England and Wales, and through October 31, 2015 for participants in Scotland. We assessed total cancer (excluding nonmelanoma skin cancer) and the 19 site-specific cancers with the highest incidence rates in the UK Biobank (≥200 incident cases), which included female breast cancer, prostate cancer, nonmelanoma skin cancer, melanoma, colorectal cancer, hematopoietic cancers, lung cancer, uterine cancer, ovarian cancer, urothelial cancer, kidney cancer, pancreatic cancer, esophageal cancer, gastric cancer, liver cancer, central nervous system cancer, mesothelioma, thyroid cancer, and head and neck cancer (HNC). Detailed ICD-10 codes for each cancer site can be found in Supplementary Table S1. The study focused on incidence cases that were developed after the initial exam date (baseline). Prevalent cases, defined as cancer diagnosed before the initial exam date, were excluded.

Statistical analyses

Reproducibility of IGF-1 over time was evaluated by calculating the intraclass correlation coefficient (ICC) among participants with IGF-1 measured at two time points. Participants were grouped into sex-specific quintiles based on their baseline IGF-1 measurements. Test for linear-trend was calculated by assigning the quintile-specific median IGF-1 value and evaluating it as a continuous variable. IGF-1 was also assessed continuously in per-5 nmol/L increments. To avoid potential biases from outlier measurements, we truncated IGF-1 measurements at 50 nmol/L. Furthermore, to avoid possible underestimation of the true association between IGF-1 and cancers due to within-person variability over time or measurement errors, we additionally corrected for regression dilution by multiplying the baseline IGF-1 by the Pearson correlation coefficients between the baseline and follow-up measurements (19, 20). This correction can provide the true strength of the association between IGF-1 and risk of cancers if there were no random fluctuations.

Cox proportional hazards models were used to calculate HR and corresponding 95% confidence interval (CI). Follow-up time was calculated from the baseline exam date until the first cancer diagnosis, death, loss to follow-up, or end of follow-up, whichever came first. We fit three separate models to examine the associations between IGF-1 and overall cancer risk and each of the 19 cancers. Model 1 included age, sex, assessment center, and the first two principal components (as a proxy for ancestry). Model 2 additionally adjusted for alcohol consumption, smoking status, physical activity, height, body mass index (BMI), waist-to-hip ratio, household income, and family history of breast, lung, colorectal, or prostate cancer (where appropriate). For female-specific cancers, we additionally adjusted in Model 2 for menopausal status, age at menarche, parity, age at first live birth, prior breast cancer screening/mammograms, and history of oral contraceptive or hormone replacement therapy use. We considered Model 2 to be the main model. Model 3 further adjusted for hsCRP, HbA1c, SHBG, and testosterone to examine whether they influenced the association between IGF-1 and individual cancer risk. In post hoc analyses, for kidney cancer, we additionally adjusted for creatinine and cystatin-C in addition to the covariates in Model 3. Similarly, for liver cancer, we additionally adjusted for ALT, AST, GGT, and total bilirubin. The proportional hazards assumption was assessed by testing an interaction term with follow-up time as well as visual examination of Schoenfeld residuals, with no evidence of deviation for any cancers. To account for potential competing risks of deaths from other diseases, in a sensitivity analysis, we performed competing risks analysis using the Fine–Gray model (21).

For cancers with large numbers of incident cases (total cancer, breast, prostate, colorectal, and lung), for which there may be biologically plausible interactions with other demographic or clinical characteristics, we conducted exploratory analysis to assess for potential effect modifiers. These included follow-up duration, sex, menopausal status, BMI, alcohol consumption, smoking status, and SHBG and testosterone levels. Heterogeneity between the subgroups was assessed with an interaction term between these factors and IGF-1.

All statistical analyses were performed using SAS 9.4 (SAS Institute) and Stata 16.1 (StataCorp). Statistical tests were two-sided and a P < 0.05 was considered statistically significant unless otherwise noted. The manuscript was prepared in accordance with STROBE (22).

Ethical approval

Ethical approval was obtained for the UK Biobank studies from the North West Multi-centre Research Ethics Committee, the National Information Governance Board for Health and Social Care in England and Wales, and the Community Health Index Advisory Group in Scotland. All participants provided written informed consent.

Data sharing

Statistical codes used for the present analysis can be made available upon request by contacting the corresponding author ([email protected]).

Participant characteristics

The baseline characteristics of 216,562 female and 196,083 male participants are shown in Table 1. In general, participants with higher circulating IGF-1 tended to be younger, taller, have a lower BMI and waist circumference, and less likely to be a smoker or daily alcohol drinker; and among females, tend to be premenopausal, nulliparous, older at first live birth, and to have ever used oral contraceptives. For circulating biomarkers, participants with higher IGF-1 tended to have lower hsCRP and SHBG. Detailed information on number of participants included in the analysis for each cancer site can be found in Supplementary Table S1.

Table 1.

Baseline characteristics of participants in the UK Biobank by sex-specific quintiles of IGF-1.

MenWomen
Quintile of IGF-1Quintile of IGF-1
Q1Q2Q3Q4Q5Q1Q2Q3Q4Q5
Participants, n 39,215 39,217 39,221 39,217 39,213 43,309 43,320 43,313 43,305 43,315 
IGF-1, median (range) 15.37 (2.02–17.57) 19.21 (17.57–20.62) 21.89 (20.62–23.13) 24.47 (23.13–26.13) 28.68 (26.13–126.77) 14.16 (1.45–16.29) 17.97 (16.29–19.47) 20.89 (19.47–22.25) 23.71 (22.25–25.49) 28.2 (25.49–125.1) 
Demographic characteristics 
 Age, years 59.5 (7.4) 58 (7.9) 56.9 (8.1) 56 (8.3) 54.6 (8.5) 60.1 (6.8) 58.5 (7.4) 57.2 (7.8) 55.7 (8) 53.5 (8.2) 
 Household income before tax, % 
  Less than £18,000 22.2 18.3 16.2 15.6 14.5 26.0 21.6 19.3 17.9 16.8 
  £18,000 to £30,999 23.9 22.3 21.5 20.6 19.9 22.5 22.6 22.2 21.6 20.5 
  £31,000 to £51,999 22.0 24.0 25.0 25.3 25.6 17.9 20.2 21.3 22.2 23.3 
  £52,000 to £100,000 15.7 19.3 21.3 22.2 23.7 10.6 13.8 15.5 17.3 19.4 
  Greater than £100,000 4.2 5.3 5.8 6.3 6.6 2.4 3.3 3.9 4.7 5.3 
  Refused 8.6 8.2 8.0 7.8 7.5 12.6 12.1 11.9 11.0 10.2 
  Do not know 3.5 2.6 2.2 2.2 2.1 7.9 6.4 5.8 5.2 4.5 
Anthropometric measurements 
 Height, cm 174.5 (6.9) 175.3 (6.8) 175.8 (6.8) 176.2 (6.8) 176.8 (6.8) 161.5 (6.3) 162.2 (6.2) 162.5 (6.2) 162.9 (6.3) 163.4 (6.3) 
 BMI, kg/m2 28.3 (4.8) 27.6 (4.1) 27.5 (3.9) 27.3 (3.7) 27.3 (3.5) 28.3 (6) 27.2 (5.2) 26.7 (4.8) 26.4 (4.5) 26.1 (4.2) 
 Waist circumference, cm 98.3 (12.4) 96.5 (10.9) 95.9 (10.5) 95.4 (10.2) 95.2 (9.8) 87.6 (13.8) 84.9 (12.4) 83.8 (11.7) 83.1 (11.2) 82.2 (10.6) 
 Hip circumference, cm 103.8 (8.6) 103.1 (7.4) 102.9 (7) 102.9 (6.8) 102.9 (6.6) 105.3 (11.8) 103.5 (10.4) 102.8 (9.7) 102.2 (9.2) 101.7 (8.7) 
 Waist-to-hip ratio 0.94 (0.07) 0.93 (0.06) 0.93 (0.06) 0.93 (0.06) 0.92 (0.06) 0.82 (0.07) 0.83 (0.07) 0.81 (0.07) 0.81 (0.07) 0.81 (0.07) 
Lifestyle factors 
 Smoking status, % 
  Never 42.9 47.5 50.4 53.1 56.1 57.6 58.7 60.3 61.2 62.7 
  Former 42.2 39.5 37.6 35.6 33.3 33.6 32.6 31.2 30.0 28.4 
  Current 15.0 13.0 12.0 11.3 10.6 8.7 8.7 8.5 8.8 8.9 
 Alcohol consumption, % 
  Never 7.2 5.6 5.5 5.4 5.7 12.4 9.2 8.3 8.1 7.9 
  Special occasions only 6.9 6.6 6.6 6.8 7.6 17.4 14.7 13.8 13.5 14.1 
  One to three times a month 7.3 8.0 8.3 9.3 10.7 12.5 12.6 12.8 13.2 14.0 
  Once or twice a week 21.7 24.4 26.1 27.6 29.7 23.2 25.1 26.1 26.9 28.6 
  Three or four times a week 24.3 27.0 27.4 27.8 27.0 18.0 20.8 21.9 22.2 21.8 
  Daily or almost daily 32.6 28.3 26.2 23.2 19.3 16.5 17.6 17.1 16.1 13.7 
 Physical activity (IPAQ scale), % 
  Low 16.5 15.4 15.0 15.4 16.0 15.0 14.0 13.8 14.0 14.0 
  Moderate 31.4 31.9 32.8 33.0 33.3 32.1 33.1 33.7 34.1 35.1 
  High 35.1 37.0 37.4 37.2 36.6 27.6 30.1 30.6 31.2 31.4 
  Missing 16.9 15.7 14.8 14.4 14.1 25.3 22.9 21.9 20.7 19.5 
Reproductive factors 
 Menopausal status, % 
  Premenopausal — — — — — 10.6 17.1 22.5 29.2 39.3 
  Postmenopausal — — — — — 86.2 79.2 73.3 66.3 55.8 
  Unknown menopausal status — — — — — 3.1 3.8 4.2 4.5 4.9 
 Age at menarche, years, % 
  <12 — — — — — 19.2 18.9 19.1 19.1 19.5 
  12 to 13 — — — — — 39.9 41.0 42.7 43.2 44.6 
  ≥14 — — — — — 37.9 37.1 35.4 34.7 32.9 
  Missing — — — — — 3.0 3.0 2.8 3.0 3.0 
 Parity, % 
  Nulliparous — — — — — 16.2 16.6 18.3 19.0 20.9 
  1 — — — — — 12.6 12.7 12.9 13.4 14.2 
  2 — — — — — 42.8 44.6 45.4 44.8 44.0 
  3 — — — — — 20.0 19.0 17.7 17.1 16.0 
  ≥4 — — — — — 8.3 7.1 5.7 5.6 4.8 
 Age at first live birth — — — — — 24.6 (4.5) 25.1 (4.5) 25.5 (4.6) 25.7 (4.6) 26.2 (4.8) 
Medical history, % 
 Family history of breast cancer in first-degree relatives — — — — — 11.5 11.3 11.3 11.1 10.9 
 Family history of lung cancer in first-degree relatives 13.6 12.9 12.2 11.4 10.5 14.4 13.1 12.7 12.1 10.8 
 Family history of prostate cancer in first-degree relatives 7.6 7.7 7.9 7.6 8.1 — — — — — 
 Family history of colorectal cancer in first-degree relatives 11.7 11.4 11.0 10.9 10.7 11.5 11.2 10.8 10.3 9.7 
 Ever used oral contraceptives — — — — — 76.0 79.1 80.4 82.6 83.5 
 Ever used oral hormone replacement therapy — — — — — 43.0 37.9 33.8 30.2 25.3 
 Ever received breast cancer screening/mammograms — — — — — 90.1 85.6 81.4 76.3 67.5 
Biomarkers 
 High-sensitivity C-reactive protein, mg/L, median (IQR) 1.69 (0.87–3.23) 1.38 (0.72–2.69) 1.24 (0.65–2.4) 1.12 (0.6–2.18) 0.98 (0.52–1.95) 2.15 (1.00–4.50) 1.53 (0.74–3.16) 1.27 (0.63–2.6) 1.08 (0.54–2.2) 0.86 (0.45–1.76) 
 Sex hormone binding globulin, nmol/L, median (IQR) 42.30 (31.89–55.5) 39.21 (29.79–50.58) 37.18 (28.39–47.85) 35.68 (27.35–45.81) 33.06 (25.22–42.55) 58.65 (40.76–81.69) 57.59 (41.24–77.87) 56.59 (40.98–75.46) 55.54 (40.56–73.66) 53.11 (39.08–70.46) 
 Testosterone, nmol/L, median (IQR) 11.80 (9.50–14.45) 11.87 (9.65–14.35) 11.75 (9.63–14.23) 11.72 (9.59–14.16) 11.56 (9.45–13.99) 0.95 (0.68–1.30) 0.99 (0.71–1.33) 1.02 (0.73–1.38) 1.05 (0.75–1.41) 1.10 (0.79–1.47) 
 HbA1c, %, median (IQR) 5.4 (5.2–5.6) 5.4 (5.1–5.6) 5.4 (5.1–5.6) 5.3 (5.1–5.6) 5.3 (5.1–5.6) 5.4 (5.2–5.6) 5.4 (5.2–5.6) 5.4 (5.2–5.6) 5.4 (5.1–5.6) 5.3 (5.1–5.5) 
MenWomen
Quintile of IGF-1Quintile of IGF-1
Q1Q2Q3Q4Q5Q1Q2Q3Q4Q5
Participants, n 39,215 39,217 39,221 39,217 39,213 43,309 43,320 43,313 43,305 43,315 
IGF-1, median (range) 15.37 (2.02–17.57) 19.21 (17.57–20.62) 21.89 (20.62–23.13) 24.47 (23.13–26.13) 28.68 (26.13–126.77) 14.16 (1.45–16.29) 17.97 (16.29–19.47) 20.89 (19.47–22.25) 23.71 (22.25–25.49) 28.2 (25.49–125.1) 
Demographic characteristics 
 Age, years 59.5 (7.4) 58 (7.9) 56.9 (8.1) 56 (8.3) 54.6 (8.5) 60.1 (6.8) 58.5 (7.4) 57.2 (7.8) 55.7 (8) 53.5 (8.2) 
 Household income before tax, % 
  Less than £18,000 22.2 18.3 16.2 15.6 14.5 26.0 21.6 19.3 17.9 16.8 
  £18,000 to £30,999 23.9 22.3 21.5 20.6 19.9 22.5 22.6 22.2 21.6 20.5 
  £31,000 to £51,999 22.0 24.0 25.0 25.3 25.6 17.9 20.2 21.3 22.2 23.3 
  £52,000 to £100,000 15.7 19.3 21.3 22.2 23.7 10.6 13.8 15.5 17.3 19.4 
  Greater than £100,000 4.2 5.3 5.8 6.3 6.6 2.4 3.3 3.9 4.7 5.3 
  Refused 8.6 8.2 8.0 7.8 7.5 12.6 12.1 11.9 11.0 10.2 
  Do not know 3.5 2.6 2.2 2.2 2.1 7.9 6.4 5.8 5.2 4.5 
Anthropometric measurements 
 Height, cm 174.5 (6.9) 175.3 (6.8) 175.8 (6.8) 176.2 (6.8) 176.8 (6.8) 161.5 (6.3) 162.2 (6.2) 162.5 (6.2) 162.9 (6.3) 163.4 (6.3) 
 BMI, kg/m2 28.3 (4.8) 27.6 (4.1) 27.5 (3.9) 27.3 (3.7) 27.3 (3.5) 28.3 (6) 27.2 (5.2) 26.7 (4.8) 26.4 (4.5) 26.1 (4.2) 
 Waist circumference, cm 98.3 (12.4) 96.5 (10.9) 95.9 (10.5) 95.4 (10.2) 95.2 (9.8) 87.6 (13.8) 84.9 (12.4) 83.8 (11.7) 83.1 (11.2) 82.2 (10.6) 
 Hip circumference, cm 103.8 (8.6) 103.1 (7.4) 102.9 (7) 102.9 (6.8) 102.9 (6.6) 105.3 (11.8) 103.5 (10.4) 102.8 (9.7) 102.2 (9.2) 101.7 (8.7) 
 Waist-to-hip ratio 0.94 (0.07) 0.93 (0.06) 0.93 (0.06) 0.93 (0.06) 0.92 (0.06) 0.82 (0.07) 0.83 (0.07) 0.81 (0.07) 0.81 (0.07) 0.81 (0.07) 
Lifestyle factors 
 Smoking status, % 
  Never 42.9 47.5 50.4 53.1 56.1 57.6 58.7 60.3 61.2 62.7 
  Former 42.2 39.5 37.6 35.6 33.3 33.6 32.6 31.2 30.0 28.4 
  Current 15.0 13.0 12.0 11.3 10.6 8.7 8.7 8.5 8.8 8.9 
 Alcohol consumption, % 
  Never 7.2 5.6 5.5 5.4 5.7 12.4 9.2 8.3 8.1 7.9 
  Special occasions only 6.9 6.6 6.6 6.8 7.6 17.4 14.7 13.8 13.5 14.1 
  One to three times a month 7.3 8.0 8.3 9.3 10.7 12.5 12.6 12.8 13.2 14.0 
  Once or twice a week 21.7 24.4 26.1 27.6 29.7 23.2 25.1 26.1 26.9 28.6 
  Three or four times a week 24.3 27.0 27.4 27.8 27.0 18.0 20.8 21.9 22.2 21.8 
  Daily or almost daily 32.6 28.3 26.2 23.2 19.3 16.5 17.6 17.1 16.1 13.7 
 Physical activity (IPAQ scale), % 
  Low 16.5 15.4 15.0 15.4 16.0 15.0 14.0 13.8 14.0 14.0 
  Moderate 31.4 31.9 32.8 33.0 33.3 32.1 33.1 33.7 34.1 35.1 
  High 35.1 37.0 37.4 37.2 36.6 27.6 30.1 30.6 31.2 31.4 
  Missing 16.9 15.7 14.8 14.4 14.1 25.3 22.9 21.9 20.7 19.5 
Reproductive factors 
 Menopausal status, % 
  Premenopausal — — — — — 10.6 17.1 22.5 29.2 39.3 
  Postmenopausal — — — — — 86.2 79.2 73.3 66.3 55.8 
  Unknown menopausal status — — — — — 3.1 3.8 4.2 4.5 4.9 
 Age at menarche, years, % 
  <12 — — — — — 19.2 18.9 19.1 19.1 19.5 
  12 to 13 — — — — — 39.9 41.0 42.7 43.2 44.6 
  ≥14 — — — — — 37.9 37.1 35.4 34.7 32.9 
  Missing — — — — — 3.0 3.0 2.8 3.0 3.0 
 Parity, % 
  Nulliparous — — — — — 16.2 16.6 18.3 19.0 20.9 
  1 — — — — — 12.6 12.7 12.9 13.4 14.2 
  2 — — — — — 42.8 44.6 45.4 44.8 44.0 
  3 — — — — — 20.0 19.0 17.7 17.1 16.0 
  ≥4 — — — — — 8.3 7.1 5.7 5.6 4.8 
 Age at first live birth — — — — — 24.6 (4.5) 25.1 (4.5) 25.5 (4.6) 25.7 (4.6) 26.2 (4.8) 
Medical history, % 
 Family history of breast cancer in first-degree relatives — — — — — 11.5 11.3 11.3 11.1 10.9 
 Family history of lung cancer in first-degree relatives 13.6 12.9 12.2 11.4 10.5 14.4 13.1 12.7 12.1 10.8 
 Family history of prostate cancer in first-degree relatives 7.6 7.7 7.9 7.6 8.1 — — — — — 
 Family history of colorectal cancer in first-degree relatives 11.7 11.4 11.0 10.9 10.7 11.5 11.2 10.8 10.3 9.7 
 Ever used oral contraceptives — — — — — 76.0 79.1 80.4 82.6 83.5 
 Ever used oral hormone replacement therapy — — — — — 43.0 37.9 33.8 30.2 25.3 
 Ever received breast cancer screening/mammograms — — — — — 90.1 85.6 81.4 76.3 67.5 
Biomarkers 
 High-sensitivity C-reactive protein, mg/L, median (IQR) 1.69 (0.87–3.23) 1.38 (0.72–2.69) 1.24 (0.65–2.4) 1.12 (0.6–2.18) 0.98 (0.52–1.95) 2.15 (1.00–4.50) 1.53 (0.74–3.16) 1.27 (0.63–2.6) 1.08 (0.54–2.2) 0.86 (0.45–1.76) 
 Sex hormone binding globulin, nmol/L, median (IQR) 42.30 (31.89–55.5) 39.21 (29.79–50.58) 37.18 (28.39–47.85) 35.68 (27.35–45.81) 33.06 (25.22–42.55) 58.65 (40.76–81.69) 57.59 (41.24–77.87) 56.59 (40.98–75.46) 55.54 (40.56–73.66) 53.11 (39.08–70.46) 
 Testosterone, nmol/L, median (IQR) 11.80 (9.50–14.45) 11.87 (9.65–14.35) 11.75 (9.63–14.23) 11.72 (9.59–14.16) 11.56 (9.45–13.99) 0.95 (0.68–1.30) 0.99 (0.71–1.33) 1.02 (0.73–1.38) 1.05 (0.75–1.41) 1.10 (0.79–1.47) 
 HbA1c, %, median (IQR) 5.4 (5.2–5.6) 5.4 (5.1–5.6) 5.4 (5.1–5.6) 5.3 (5.1–5.6) 5.3 (5.1–5.6) 5.4 (5.2–5.6) 5.4 (5.2–5.6) 5.4 (5.2–5.6) 5.4 (5.1–5.6) 5.3 (5.1–5.5) 

Note: Data are mean (SD) unless otherwise indicated.

Abbreviations: IPAQ, International Physical Activity Questionnaire; IQR, interquartile range; SD, standard deviation.

IGF-1 and total and site-specific cancer risks

IGF-1 levels measured at two time points (∼4 years apart) in a subset of participants was highly reproducible over time (ICC: 0.75, Pearson correlation coefficient: 0.77). Regression dilution was corrected by multiplying the measured IGF-1 values by 0.77.

Participants were followed for a median of 7.2 years. Higher IGF-1 was associated with increased total cancer risk in both men [HR (95% CI) comparing Q5 vs. Q1: 1.08 (1.02–1.15)] and women [1.08 (1.02–1.15)]. For site-specific cancers, higher IGF-1 was associated with an increased risk of cancer of the breast (HR Q5 vs. Q1: 1.24; 95% CI, 1.12–1.36), prostate 1.20 (1.10–1.30), colorectum 1.21 (1.08–1.36), kidney 1.29 (1.01–1.64), and thyroid 1.50 (0.97–2.32; Table 2). Conversely, higher circulating IGF-1 was associated with lower risks of lung 0.84 (0.72–0.97), ovarian 0.64 (0.47–0.87), and liver cancers 0.21 (0.13–0.36). The inverse association for lung cancer was only observed among ever-smokers (1,695 cases): 0.79 (0.67, 0.93), whereas among never-smokers (301 cases), the association trended toward increased risk 1.22 (0.84, 1.76). Further adjustment for hsCRP, HbA1c, SHBG, and testosterone did not materially alter these associations (Model 3 in Supplementary Tables S2 and S3). We did not find appreciable associations between IGF-1 and the other cancer sites we evaluated.

Table 2.

Association of circulating IGF-1 with incident total and site-specific cancersa.

Quintile of IGF-1
Q1Q2Q3Q4Q5Ptrend
Total cancer,bwomen (N = 11,137) 
Cases/person-years 2,471/278,733 2,335/283,919 2,212/284,491 2,123/285,907 1,996/286,424  
 Model 1, HR (95% CI) 1.00 (ref.) 0.99 (0.93–1.05) 0.99 (0.93–1.04) 1.00 (0.95–1.06) 1.04 (0.98–1.10) 0.23 
 Model 2, HR (95% CI) 1.00 (ref.) 1.01 (0.95–1.07) 1.02 (0.96–1.08) 1.04 (0.98–1.10) 1.08 (1.02–1.15) 0.008 
Total cancer,bmen (N = 12,854) 
Cases/person-years 3,037/256,324 2,728/259,529 2,494/260,084 2,382/259,730 2,213/259,557  
 Model 1, HR (95% CI) 1.00 (ref.) 0.99 (0.94–1.04) 0.99 (0.93–1.04) 1.00 (0.95–1.06) 1.05 (1.00–1.11) 0.09 
 Model 2, HR (95% CI) 1.00 (ref.) 1.00 (0.95–1.06) 1.01 (0.95–1.06) 1.03 (0.98–1.09) 1.08 (1.02–1.15) 0.005 
Breast cancer (N = 4,666) 
Cases/person-years 901/289,734 884/294,022 966/294,238 943/295,019 972/295,111  
 Model 1, HR (95% CI) 1.00 (ref.) 1.00 (0.91–1.09) 1.12 (1.02–1.23) 1.13 (1.03–1.23) 1.22 (1.11–1.34) <0.001 
 Model 2, HR (95% CI) 1.00 (ref.) 1.00 (0.91–1.10) 1.13 (1.03–1.24) 1.13 (1.03–1.25) 1.24 (1.12–1.36) <0.001 
Prostate cancer (N = 5,548) 
Cases/person-years 1,214/266,774 1,195/269,067 1,114/269,306 1,034/269,280 991/268,868  
 Model 1, HR (95% CI) 1.00 (ref.) 1.10 (1.02–1.19) 1.13 (1.04–1.23) 1.13 (1.04–1.23) 1.24 (1.14–1.35) <0.001 
 Model 2, HR (95% CI) 1.00 (ref.) 1.08 (0.99–1.17) 1.10 (1.01–1.19) 1.10 (1.01–1.19) 1.20 (1.10–1.30) <0.001 
Colorectal cancer (N = 2,975) 
Cases/person-years 666/575,984 656/579,238 591/579,403 528/578,508 534/577,838  
 Model 1, HR (95% CI) 1.00 (ref.) 1.08 (0.97–1.20) 1.05 (0.94–1.17) 1.02 (0.91–1.14) 1.17 (1.04–1.31) 0.04 
 Model 2, HR (95% CI) 1.00 (ref.) 1.10 (0.98–1.22) 1.08 (0.96–1.21) 1.05 (0.94–1.18) 1.21 (1.08–1.36) 0.007 
Nonmelanoma skin cancer (N = 11,879) 
Cases/person-years 2,771/558,945 2,531/564,215 2,466/564,584 2,185/565,210 1,926/566,431  
 Model 1, HR (95% CI) 1.00 (ref.) 0.99 (0.94–1.05) 1.04 (0.99–1.10) 1.00 (0.94–1.06) 0.99 (0.94–1.05) 0.97 
 Model 2, HR (95% CI) 1.00 (ref.) 0.96 (0.91–1.01) 0.99 (0.94–1.05) 0.95 (0.89–1.00) 0.94 (0.88–1.00) 0.04 
Melanoma (N = 1,502) 
Cases/person-years 300/577,378 311/580,780 295/580,129 295/579,507 301/578,746  
 Model 1, HR (95% CI) 1.00 (ref.) 1.08 (0.92–1.27) 1.07 (0.91–1.26) 1.12 (0.96–1.32) 1.23 (1.05–1.45) 0.01 
 Model 2, HR (95% CI) 1.00 (ref.) 1.04 (0.89–1.23) 1.02 (0.87–1.20) 1.06 (0.90–1.25) 1.15 (0.98–1.36) 0.09 
Lung cancer (N = 1,996) 
Cases/person-years 591/579,270 468/582,831 354/582,747 323/581,875 260/581,183  
 Model 1, HR (95% CI) 1.00 (ref.) 0.89 (0.78–1.00) 0.74 (0.65–0.84) 0.74 (0.65–0.85) 0.70 (0.60–0.81) <0.0001 
 Model 2, HR (95% CI) 1.00 (ref.) 0.96 (0.85–1.09) 0.85 (0.74–0.97) 0.87 (0.76–1.00) 0.84 (0.72–0.97) 0.005 
Hematopoietic cancer (N = 2,310) 
Cases/person-years 563/576,726 490/580,507 426/580,368 425/579,320 406/578,671  
 Model 1, HR (95% CI) 1.00 (ref.) 0.95 (0.84–1.07) 0.89 (0.78–1.00) 0.95 (0.84–1.08) 1.02 (0.90–1.16) 0.90 
 Model 2, HR (95% CI) 1.00 (ref.) 0.95 (0.84–1.07) 0.89 (0.78–1.01) 0.95 (0.84–1.08) 1.01 (0.89–1.16) 0.98 
Uterine cancer (N = 777) 
Cases/person-years 205/304,209 181/305,150 145/305,097 134/304,859 112/304,914  
 Model 1, HR (95% CI) 1.00 (ref.) 0.94 (0.77–1.15) 0.81 (0.65–1.00) 0.80 (0.64–0.99) 0.76 (0.60–0.96) 0.006 
 Model 2, HR (95% CI) 1.00 (ref.) 1.05 (0.86–1.29) 0.93 (0.75–1.16) 0.95 (0.76–1.18) 0.93 (0.73–1.18) 0.38 
Kidney cancer (N = 691) 
Cases/person-years 156/579,720 137/582,969 131/582,658 139/581,614 128/580,641  
 Model 1, HR (95% CI) 1.00 (ref.) 0.95 (0.76–1.20) 0.98 (0.78–1.24) 1.12 (0.89–1.42) 1.17 (0.92–1.48) 0.11 
 Model 2, HR (95% CI) 1.00 (ref.) 1.02 (0.81–1.28) 1.08 (0.85–1.36) 1.24 (0.98–1.56) 1.29 (1.01–1.64) 0.02 
Urothelial cancer (N = 677) 
Cases/person-years 173/579,213 145/582,637 127/582,641 111/581,590 121/580,793  
 Model 1, HR (95% CI) 1.00 (ref.) 0.93 (0.75–1.17) 0.90 (0.72–1.14) 0.87 (0.68–1.10) 1.10 (0.87–1.40) 0.72 
 Model 2, HR (95% CI) 1.00 (ref.) 0.97 (0.78–1.21) 0.96 (0.76–1.21) 0.93 (0.73–1.19) 1.20 (0.95–1.52) 0.27 
HNC (N = 665) 
Cases/person-years 182/578,861 139/582,650 132/582,308 109/581,417 103/580,855  
 Model 1, HR (95% CI) 1.00 (ref.) 0.79 (0.64–0.99) 0.78 (0.62–0.98) 0.67 (0.53–0.85) 0.68 (0.53–0.87) 0.001 
 Model 2, HR (95% CI) 1.00 (ref.) 0.87 (0.70–1.09) 0.90 (0.72–1.13) 0.80 (0.63–1.02) 0.85 (0.66–1.08) 0.12 
Ovarian cancer (N = 532) 
Cases/person-years 129/305,001 134/305,942 104/305,484 99/305,470 66/305,313  
 Model 1, HR (95% CI) 1.00 (ref.) 1.10 (0.86–1.40) 0.90 (0.69–1.17) 0.91 (0.70–1.18) 0.66 (0.48–0.89) 0.004 
 Model 2, HR (95% CI) 1.00 (ref.) 1.10 (0.86–1.41) 0.89 (0.69–1.16) 0.90 (0.69–1.17) 0.64 (0.47–0.87) 0.002 
Pancreatic cancer (N = 569) 
Cases/person-years 136/580,292 114/583,588 106/583,323 127/582,394 86/581,678  
 Model 1, HR (95% CI) 1.00 (ref.) 0.93 (0.73–1.19) 0.94 (0.73–1.22) 1.24 (0.97–1.59) 0.98 (0.75–1.29) 0.47 
 Model 2, HR (95% CI) 1.00 (ref.) 0.97 (0.75–1.24) 1.00 (0.77–1.29) 1.32 (1.04–1.69) 1.05 (0.80–1.39) 0.15 
Esophageal cancer (N = 496) 
Cases/person-years 154/580,003 96/583,426 82/583,216 91/582,242 73/581,465  
 Model 1, HR (95% CI) 1.00 (ref.) 0.69 (0.54–0.89) 0.65 (0.49–0.85) 0.78 (0.60–1.02) 0.73 (0.55–0.97) 0.03 
 Model 2, HR (95% CI) 1.00 (ref.) 0.75 (0.58–0.97) 0.72 (0.55–0.95) 0.90 (0.69–1.17) 0.84 (0.63–1.12) 0.29 
Central nervous system cancer (N = 436) 
Cases/person-years 96/580,257 81/583,560 87/583,267 88/582,381 84/581,507  
 Model 1, HR (95% CI) 1.00 (ref.) 0.90 (0.67–1.21) 1.02 (0.76–1.37) 1.10 (0.82–1.47) 1.16 (0.86–1.56) 0.17 
 Model 2, HR (95% CI) 1.00 (ref.) 0.88 (0.66–1.19) 1.00 (0.75–1.34) 1.07 (0.80–1.44) 1.13 (0.83–1.53) 0.24 
Gastric cancer (N = 334) 
Cases/person-years 87/580,067 73/583,384 72/583,273 55/582,303 47/581,637  
 Model 1, HR (95% CI) 1.00 (ref.) 0.92 (0.68–1.26) 0.98 (0.72–1.35) 0.81 (0.58–1.14) 0.78 (0.54–1.12) 0.14 
 Model 2, HR (95% CI) 1.00 (ref.) 0.99 (0.72–1.35) 1.08 (0.79–1.48) 0.90 (0.64–1.27) 0.86 (0.60–1.24) 0.42 
Liver cancer (N = 238) 
Cases/person-years 123/580,259 47/583,635 24/583,448 27/582,497 17/581,694  
 Model 1, HR (95% CI) 1.00 (ref.) 0.41 (0.29–0.58) 0.22 (0.14–0.35) 0.27 (0.18–0.41) 0.19 (0.11–0.32) <0.001 
 Model 2, HR (95% CI) 1.00 (ref.) 0.44 (0.32–0.62) 0.25 (0.16–0.38) 0.30 (0.20–0.46) 0.21 (0.13–0.36) <0.001 
Mesothelioma (N = 240) 
Cases/person-years 58/580,351 46/583,669 48/583,415 48/582,477 40/581,709  
 Model 1, HR (95% CI) 1.00 (ref.) 0.91 (0.62–1.34) 1.07 (0.73–1.57) 1.20 (0.81–1.76) 1.18 (0.79–1.77) 0.23 
 Model 2, HR (95% CI) 1.00 (ref.) 0.91 (0.62–1.34) 1.08 (0.73–1.59) 1.21 (0.82–1.77) 1.18 (0.78–1.78) 0.23 
Thyroid cancer (N = 231) 
Cases/person-years 38/579,913 37/583,230 45/582,945 59/582,025 52/581,146  
 Model 1, HR (95% CI) 1.00 (ref.) 0.99 (0.63–1.56) 1.22 (0.79–1.88) 1.63 (1.08–2.46) 1.48 (0.96–2.27) 0.01 
 Model 2, HR (95% CI) 1.00 (ref.) 1.00 (0.64–1.58) 1.24 (0.80–1.92) 1.66 (1.09–2.51) 1.50 (0.97–2.32) 0.01 
Quintile of IGF-1
Q1Q2Q3Q4Q5Ptrend
Total cancer,bwomen (N = 11,137) 
Cases/person-years 2,471/278,733 2,335/283,919 2,212/284,491 2,123/285,907 1,996/286,424  
 Model 1, HR (95% CI) 1.00 (ref.) 0.99 (0.93–1.05) 0.99 (0.93–1.04) 1.00 (0.95–1.06) 1.04 (0.98–1.10) 0.23 
 Model 2, HR (95% CI) 1.00 (ref.) 1.01 (0.95–1.07) 1.02 (0.96–1.08) 1.04 (0.98–1.10) 1.08 (1.02–1.15) 0.008 
Total cancer,bmen (N = 12,854) 
Cases/person-years 3,037/256,324 2,728/259,529 2,494/260,084 2,382/259,730 2,213/259,557  
 Model 1, HR (95% CI) 1.00 (ref.) 0.99 (0.94–1.04) 0.99 (0.93–1.04) 1.00 (0.95–1.06) 1.05 (1.00–1.11) 0.09 
 Model 2, HR (95% CI) 1.00 (ref.) 1.00 (0.95–1.06) 1.01 (0.95–1.06) 1.03 (0.98–1.09) 1.08 (1.02–1.15) 0.005 
Breast cancer (N = 4,666) 
Cases/person-years 901/289,734 884/294,022 966/294,238 943/295,019 972/295,111  
 Model 1, HR (95% CI) 1.00 (ref.) 1.00 (0.91–1.09) 1.12 (1.02–1.23) 1.13 (1.03–1.23) 1.22 (1.11–1.34) <0.001 
 Model 2, HR (95% CI) 1.00 (ref.) 1.00 (0.91–1.10) 1.13 (1.03–1.24) 1.13 (1.03–1.25) 1.24 (1.12–1.36) <0.001 
Prostate cancer (N = 5,548) 
Cases/person-years 1,214/266,774 1,195/269,067 1,114/269,306 1,034/269,280 991/268,868  
 Model 1, HR (95% CI) 1.00 (ref.) 1.10 (1.02–1.19) 1.13 (1.04–1.23) 1.13 (1.04–1.23) 1.24 (1.14–1.35) <0.001 
 Model 2, HR (95% CI) 1.00 (ref.) 1.08 (0.99–1.17) 1.10 (1.01–1.19) 1.10 (1.01–1.19) 1.20 (1.10–1.30) <0.001 
Colorectal cancer (N = 2,975) 
Cases/person-years 666/575,984 656/579,238 591/579,403 528/578,508 534/577,838  
 Model 1, HR (95% CI) 1.00 (ref.) 1.08 (0.97–1.20) 1.05 (0.94–1.17) 1.02 (0.91–1.14) 1.17 (1.04–1.31) 0.04 
 Model 2, HR (95% CI) 1.00 (ref.) 1.10 (0.98–1.22) 1.08 (0.96–1.21) 1.05 (0.94–1.18) 1.21 (1.08–1.36) 0.007 
Nonmelanoma skin cancer (N = 11,879) 
Cases/person-years 2,771/558,945 2,531/564,215 2,466/564,584 2,185/565,210 1,926/566,431  
 Model 1, HR (95% CI) 1.00 (ref.) 0.99 (0.94–1.05) 1.04 (0.99–1.10) 1.00 (0.94–1.06) 0.99 (0.94–1.05) 0.97 
 Model 2, HR (95% CI) 1.00 (ref.) 0.96 (0.91–1.01) 0.99 (0.94–1.05) 0.95 (0.89–1.00) 0.94 (0.88–1.00) 0.04 
Melanoma (N = 1,502) 
Cases/person-years 300/577,378 311/580,780 295/580,129 295/579,507 301/578,746  
 Model 1, HR (95% CI) 1.00 (ref.) 1.08 (0.92–1.27) 1.07 (0.91–1.26) 1.12 (0.96–1.32) 1.23 (1.05–1.45) 0.01 
 Model 2, HR (95% CI) 1.00 (ref.) 1.04 (0.89–1.23) 1.02 (0.87–1.20) 1.06 (0.90–1.25) 1.15 (0.98–1.36) 0.09 
Lung cancer (N = 1,996) 
Cases/person-years 591/579,270 468/582,831 354/582,747 323/581,875 260/581,183  
 Model 1, HR (95% CI) 1.00 (ref.) 0.89 (0.78–1.00) 0.74 (0.65–0.84) 0.74 (0.65–0.85) 0.70 (0.60–0.81) <0.0001 
 Model 2, HR (95% CI) 1.00 (ref.) 0.96 (0.85–1.09) 0.85 (0.74–0.97) 0.87 (0.76–1.00) 0.84 (0.72–0.97) 0.005 
Hematopoietic cancer (N = 2,310) 
Cases/person-years 563/576,726 490/580,507 426/580,368 425/579,320 406/578,671  
 Model 1, HR (95% CI) 1.00 (ref.) 0.95 (0.84–1.07) 0.89 (0.78–1.00) 0.95 (0.84–1.08) 1.02 (0.90–1.16) 0.90 
 Model 2, HR (95% CI) 1.00 (ref.) 0.95 (0.84–1.07) 0.89 (0.78–1.01) 0.95 (0.84–1.08) 1.01 (0.89–1.16) 0.98 
Uterine cancer (N = 777) 
Cases/person-years 205/304,209 181/305,150 145/305,097 134/304,859 112/304,914  
 Model 1, HR (95% CI) 1.00 (ref.) 0.94 (0.77–1.15) 0.81 (0.65–1.00) 0.80 (0.64–0.99) 0.76 (0.60–0.96) 0.006 
 Model 2, HR (95% CI) 1.00 (ref.) 1.05 (0.86–1.29) 0.93 (0.75–1.16) 0.95 (0.76–1.18) 0.93 (0.73–1.18) 0.38 
Kidney cancer (N = 691) 
Cases/person-years 156/579,720 137/582,969 131/582,658 139/581,614 128/580,641  
 Model 1, HR (95% CI) 1.00 (ref.) 0.95 (0.76–1.20) 0.98 (0.78–1.24) 1.12 (0.89–1.42) 1.17 (0.92–1.48) 0.11 
 Model 2, HR (95% CI) 1.00 (ref.) 1.02 (0.81–1.28) 1.08 (0.85–1.36) 1.24 (0.98–1.56) 1.29 (1.01–1.64) 0.02 
Urothelial cancer (N = 677) 
Cases/person-years 173/579,213 145/582,637 127/582,641 111/581,590 121/580,793  
 Model 1, HR (95% CI) 1.00 (ref.) 0.93 (0.75–1.17) 0.90 (0.72–1.14) 0.87 (0.68–1.10) 1.10 (0.87–1.40) 0.72 
 Model 2, HR (95% CI) 1.00 (ref.) 0.97 (0.78–1.21) 0.96 (0.76–1.21) 0.93 (0.73–1.19) 1.20 (0.95–1.52) 0.27 
HNC (N = 665) 
Cases/person-years 182/578,861 139/582,650 132/582,308 109/581,417 103/580,855  
 Model 1, HR (95% CI) 1.00 (ref.) 0.79 (0.64–0.99) 0.78 (0.62–0.98) 0.67 (0.53–0.85) 0.68 (0.53–0.87) 0.001 
 Model 2, HR (95% CI) 1.00 (ref.) 0.87 (0.70–1.09) 0.90 (0.72–1.13) 0.80 (0.63–1.02) 0.85 (0.66–1.08) 0.12 
Ovarian cancer (N = 532) 
Cases/person-years 129/305,001 134/305,942 104/305,484 99/305,470 66/305,313  
 Model 1, HR (95% CI) 1.00 (ref.) 1.10 (0.86–1.40) 0.90 (0.69–1.17) 0.91 (0.70–1.18) 0.66 (0.48–0.89) 0.004 
 Model 2, HR (95% CI) 1.00 (ref.) 1.10 (0.86–1.41) 0.89 (0.69–1.16) 0.90 (0.69–1.17) 0.64 (0.47–0.87) 0.002 
Pancreatic cancer (N = 569) 
Cases/person-years 136/580,292 114/583,588 106/583,323 127/582,394 86/581,678  
 Model 1, HR (95% CI) 1.00 (ref.) 0.93 (0.73–1.19) 0.94 (0.73–1.22) 1.24 (0.97–1.59) 0.98 (0.75–1.29) 0.47 
 Model 2, HR (95% CI) 1.00 (ref.) 0.97 (0.75–1.24) 1.00 (0.77–1.29) 1.32 (1.04–1.69) 1.05 (0.80–1.39) 0.15 
Esophageal cancer (N = 496) 
Cases/person-years 154/580,003 96/583,426 82/583,216 91/582,242 73/581,465  
 Model 1, HR (95% CI) 1.00 (ref.) 0.69 (0.54–0.89) 0.65 (0.49–0.85) 0.78 (0.60–1.02) 0.73 (0.55–0.97) 0.03 
 Model 2, HR (95% CI) 1.00 (ref.) 0.75 (0.58–0.97) 0.72 (0.55–0.95) 0.90 (0.69–1.17) 0.84 (0.63–1.12) 0.29 
Central nervous system cancer (N = 436) 
Cases/person-years 96/580,257 81/583,560 87/583,267 88/582,381 84/581,507  
 Model 1, HR (95% CI) 1.00 (ref.) 0.90 (0.67–1.21) 1.02 (0.76–1.37) 1.10 (0.82–1.47) 1.16 (0.86–1.56) 0.17 
 Model 2, HR (95% CI) 1.00 (ref.) 0.88 (0.66–1.19) 1.00 (0.75–1.34) 1.07 (0.80–1.44) 1.13 (0.83–1.53) 0.24 
Gastric cancer (N = 334) 
Cases/person-years 87/580,067 73/583,384 72/583,273 55/582,303 47/581,637  
 Model 1, HR (95% CI) 1.00 (ref.) 0.92 (0.68–1.26) 0.98 (0.72–1.35) 0.81 (0.58–1.14) 0.78 (0.54–1.12) 0.14 
 Model 2, HR (95% CI) 1.00 (ref.) 0.99 (0.72–1.35) 1.08 (0.79–1.48) 0.90 (0.64–1.27) 0.86 (0.60–1.24) 0.42 
Liver cancer (N = 238) 
Cases/person-years 123/580,259 47/583,635 24/583,448 27/582,497 17/581,694  
 Model 1, HR (95% CI) 1.00 (ref.) 0.41 (0.29–0.58) 0.22 (0.14–0.35) 0.27 (0.18–0.41) 0.19 (0.11–0.32) <0.001 
 Model 2, HR (95% CI) 1.00 (ref.) 0.44 (0.32–0.62) 0.25 (0.16–0.38) 0.30 (0.20–0.46) 0.21 (0.13–0.36) <0.001 
Mesothelioma (N = 240) 
Cases/person-years 58/580,351 46/583,669 48/583,415 48/582,477 40/581,709  
 Model 1, HR (95% CI) 1.00 (ref.) 0.91 (0.62–1.34) 1.07 (0.73–1.57) 1.20 (0.81–1.76) 1.18 (0.79–1.77) 0.23 
 Model 2, HR (95% CI) 1.00 (ref.) 0.91 (0.62–1.34) 1.08 (0.73–1.59) 1.21 (0.82–1.77) 1.18 (0.78–1.78) 0.23 
Thyroid cancer (N = 231) 
Cases/person-years 38/579,913 37/583,230 45/582,945 59/582,025 52/581,146  
 Model 1, HR (95% CI) 1.00 (ref.) 0.99 (0.63–1.56) 1.22 (0.79–1.88) 1.63 (1.08–2.46) 1.48 (0.96–2.27) 0.01 
 Model 2, HR (95% CI) 1.00 (ref.) 1.00 (0.64–1.58) 1.24 (0.80–1.92) 1.66 (1.09–2.51) 1.50 (0.97–2.32) 0.01 

aModel 1 adjusted for age (5-year categories), assessment center, and the first two principal components (as a proxy for genetic ancestry). Model 2 additionally adjusted for alcohol consumption (never, special occasions only, 1–3 times per month, 1–2 times per week, 3–4 times per week, daily/almost daily), smoking status (never, former, current), physical activity on the International Physical Activity Questionnaire (IPAQ) scale (low, moderate, high, missing), height in cm (women: <155, 155–159, 160–164, 165–169, ≥170; men: <165, 165–169, 170–174, 175–179, ≥180), BMI in kg/m2 (<21, 21–24.9, 25–29.9, 30–34.9, ≥35), waist-to-hip ratio (women: ≤0.80, 0.81–0.85, >0.85; men: ≤0.90, 0.91–0.95, >0.95), and average household income (less than £18,000, £18,000 to £30,999, £31,000 to £51,999, £52,000 to £100,000, greater than £100,000, refused, do not know). For female-specific cancers, we additionally adjusted for menopausal status (premenopausal, postmenopausal, unknown menopausal status), age at menarche (<12, 12–13, ≥14, missing), parity (nulliparous, 1, 2, 3, ≥4), age at first live birth (nulliparous, <20, 20–24, 25–29, ≥30), had prior breast cancer screening/mammograms (ever, never), ever used oral contraceptives (yes, no), and ever used hormone replacement therapy (yes, no). For breast, prostate, colorectal, and lung cancer, we additionally adjusted for family history of the respective cancer in a first-degree relative.

bTotal cancer excludes nonmelanoma skin cancer.

When IGF-1 was modeled continuously, we observed similar associations (Fig. 1). For a 5-nmol/L increment in IGF-1 levels that was corrected for regression dilution, we found an increased risk of total cancer [HR (95% CI), men: 1.03 (1.01–1.06); women: 1.03 (1.01–1.06)], breast 1.10 (1.07–1.14), prostate 1.09 (1.05–1.12), colorectal 1.07 (1.02–1.11), kidney 1.10 (1.00–1.20), and thyroid cancers 1.22 (1.05–1.42); and a decreased risk of lung 0.91 (0.86–0.96), ovarian 0.86 (0.77–0.95), and liver cancers 0.32 (0.26–0.38). The positive associations we observed for total cancer in women appeared to be driven by breast cancer whereas the positive association for total cancer in men appeared to be driven by prostate cancer. When we excluded these cancers from the definition of total cancer, the associations were attenuated. For total cancer in women, the HR (95% CI) per 5-nmol/L increment in IGF-1 was 0.98 (0.96–1.01); for total cancer in men, the association was 0.99 (0.97–1.02). We also observed significant associations for HNC: 0.90 (0.82–0.99) and melanoma: 1.08 (1.01–1.15), which were not statistically significant in the quintile analyses. After stringent Bonferroni adjustment (P < 0.05/21), the associations for breast, prostate, colorectal, lung, and liver cancers remained statistically significant. Associations were similar for IGF-1 that was not corrected for regression dilution (Supplementary Table S4). Finally, we performed competing risks analysis using the Fine–Gray model and found nearly identical results to those obtained using Cox models (Supplementary Table S5).

Figure 1.

Associations between IGF-1 (per 5-nmol/L increment) and total and site-specific cancers. Associations were adjusted for age (5-year categories), assessment center, the first two principal components, sex (for cancer in both men and women), alcohol consumption (never, special occasions only, 1–3 times per month, 1–2 times per week, 3–4 times per week, daily/almost daily), smoking status (never, former, current), physical activity on the International Physical Activity Questionnaire (IPAQ) scale (low, moderate, high, missing), height in cm (women: <155, 155–159, 160–164, 165–169, ≥170; men: <165, 165–169, 170–174, 175–179, ≥180), BMI in kg/m2 (<21, 21–24.9, 25–29.9, 30–34.9, ≥35), waist-to-hip ratio (women: ≤0.80, 0.81–0.85, >0.85; men: ≤0.90, 0.91–0.95, >0.95), and average household income (less than £18,000, £18,000 to £30,999, £31,000 to £51,999, £52,000 to £100,000, greater than £100,000, refused, do not know). For female-specific cancers, we additionally adjusted for menopausal status (premenopausal, postmenopausal, unknown menopausal status), age at menarche (<12, 12–13, ≥14, missing), parity (nulliparous, 1, 2, 3, ≥4), age at first live birth (nulliparous, <20, 20–24, 25–29, ≥30), ever used oral contraceptives (yes, no), and ever used hormone replacement therapy (yes, no). For breast, prostate, colorectal, and lung cancer, we additionally adjusted for family history of the respective cancer in a first-degree relative (yes, no).

Figure 1.

Associations between IGF-1 (per 5-nmol/L increment) and total and site-specific cancers. Associations were adjusted for age (5-year categories), assessment center, the first two principal components, sex (for cancer in both men and women), alcohol consumption (never, special occasions only, 1–3 times per month, 1–2 times per week, 3–4 times per week, daily/almost daily), smoking status (never, former, current), physical activity on the International Physical Activity Questionnaire (IPAQ) scale (low, moderate, high, missing), height in cm (women: <155, 155–159, 160–164, 165–169, ≥170; men: <165, 165–169, 170–174, 175–179, ≥180), BMI in kg/m2 (<21, 21–24.9, 25–29.9, 30–34.9, ≥35), waist-to-hip ratio (women: ≤0.80, 0.81–0.85, >0.85; men: ≤0.90, 0.91–0.95, >0.95), and average household income (less than £18,000, £18,000 to £30,999, £31,000 to £51,999, £52,000 to £100,000, greater than £100,000, refused, do not know). For female-specific cancers, we additionally adjusted for menopausal status (premenopausal, postmenopausal, unknown menopausal status), age at menarche (<12, 12–13, ≥14, missing), parity (nulliparous, 1, 2, 3, ≥4), age at first live birth (nulliparous, <20, 20–24, 25–29, ≥30), ever used oral contraceptives (yes, no), and ever used hormone replacement therapy (yes, no). For breast, prostate, colorectal, and lung cancer, we additionally adjusted for family history of the respective cancer in a first-degree relative (yes, no).

Close modal

Stratified and sensitivity analyses

Associations for IGF-1 were largely consistent across subgroups for total, breast, prostate, colorectal, and lung cancers (Table 3; Supplementary Fig. S1). In particular, other circulating hormones such as testosterone or SHBG did not modify the association between IGF-1 and incident breast or prostate cancer (Pint > 0.05 for both cancers). The association between IGF-1 and total cancer in men was stronger among never-smokers [HR (95% CI) per 5-nmol/L increment in IGF-1: 1.08 (1.05–1.12)] compared with ever-smokers [1.00 (0.97–1.03); Pint = 0.0003]. This appeared to be driven by heterogeneity in the association for lung cancer, for which the association was 1.14 (0.99–1.30) among never-smokers and 0.88 (0.83–0.93) among ever-smokers, Pint = 0.0005. Adjusting for smoking intensity (pack-years) did not alter the association among ever-smokers: 0.89 (0.83–0.95). In addition, the association between IGF-1 and lung cancer was only found for men [0.85 (0.79–0.91)] but not for women [1.00 (0.92–1.08); Pint = 0.003]. Both interaction terms remained significant when they were simultaneously included in the same model, Pint for ever-smoking = 0.002 and Pint for sex = 0.009.

Table 3.

Subgroup analysis of association for per 5-nmol/L increment in IGF-1 and total and selected site-specific cancers.

Total cancer (women)PhetTotal cancer (men)PhetBreast cancerPhetProstate cancerPhetColorectal cancerPhetLung cancerPhet
Overall 1.03 (1.01–1.06)  1.03 (1.01–1.06)  1.10 (1.07–1.14)  1.09 (1.05–1.12)  1.07 (1.02–1.11)  0.91 (0.86–0.96)  
Sex          0.23  0.003 
 Male —  1.03 (1.01–1.06)  —  1.09 (1.05–1.12)  1.10 (1.03–1.17)  0.85 (0.79–0.91)  
 Female 1.03 (1.01–1.06)  —  1.10 (1.07–1.14)  —  1.04 (0.98, 1.10)  1.00 (0.92–1.08)  
Menopausal status, among women only  0.07    0.06    0.28  0.29 
 Premenopausal 1.08 (1.02–1.14)  —  1.17 (1.08–1.26)  —  1.19 (0.97–1.46)  0.80 (0.54–1.19)  
 Postmenopausal 1.02 (0.99–1.04)  —  1.08 (1.03–1.12)  —  1.06 (0.99–1.14)  0.99 (0.91–1.08)  
BMI, kg/m2  0.30  0.10  0.59  0.25  1.00  0.33 
 <25 1.01 (0.98–1.05)  1.00 (0.96–1.05)  1.12 (1.05–1.18)  1.05 (0.98–1.12)  1.07 (0.98–1.16)  0.88 (0.80–0.97)  
 ≥25 1.04 (1.01–1.07)  1.04 (1.02–1.07)  1.10 (1.05–1.14)  1.10 (1.06–1.14)  1.07 (1.01–1.12)  0.93 (0.87–0.997)  
Smoking status  0.58  0.0003  0.81  0.72  0.89  0.0005 
 Never-smoker 1.04 (1.01–1.07)  1.08 (1.05–1.12)  1.10 (1.05–1.15)  1.08 (1.00–1.15)  1.07 (1.00–1.14)  1.14 (0.99–1.30)  
 Ever-smoker 1.02 (0.99–1.06)  1.00 (0.97–1.03)  1.11 (1.05–1.17)  1.09 (1.05–1.13)  1.06 (1.00–1.13)  0.88 (0.83–0.93)  
Alcohol consumption  0.84  0.23  0.37  0.89  0.87  0.54 
 Less than one drink per week 1.03 (0.99–1.06)  1.01 (0.96–1.05)  1.08 (1.02–1.14)  1.09 (1.03–1.14)  1.07 (0.99–1.16)  0.94 (0.85–1.03)  
 One drink or more per week 1.03 (1.00–1.06)  1.04 (1.01–1.07)  1.12 (1.07–1.17)  1.09 (1.04–1.14)  1.06 (1.01–1.12)  0.90 (0.84–0.97)  
Follow-up duration, years  0.43  0.45  0.56  0.24  0.60  0.82 
 <5 1.02 (1.00–1.05)  1.04 (1.01–1.07)  1.10 (1.05–1.14)  1.07 (1.03–1.12)  1.06 (1.00–1.12)  0.91 (0.85–0.98)  
 ≥5 1.04 (1.00–1.08)  1.02 (0.98–1.06)  1.12 (1.05–1.19)  1.12 (1.06–1.18)  1.08 (1.00–1.17)  0.92 (0.84–1.01)  
Sex hormone binding globulin, nmol/L  0.92  0.40  0.21  0.45  0.29  0.19 
 <Median 1.03 (1.00–1.06)  1.04 (1.01–1.08)  1.11 (1.06–1.17)  1.07 (1.01–1.12)  1.10 (1.03–1.17)  0.95 (0.87–1.04)  
 ≥Median 1.03 (0.99–1.06)  1.02 (0.99–1.05)  1.06 (1.01–1.12)  1.09 (1.04–1.15)  1.04 (0.98–1.11)  0.88 (0.82–0.95)  
Testosterone, nmol/L  0.47  0.66  0.72  0.53  0.47  0.26 
 <Median 1.02 (0.98–1.06)  1.04 (1.01–1.07)  1.08 (1.02–1.14)  1.08 (1.03–1.13)  1.05 (0.98–1.11)  0.92 (0.85–1.00)  
 ≥Median 1.04 (1.01–1.07)  1.03 (1.00–1.06)  1.09 (1.04–1.15)  1.10 (1.05–1.15)  1.08 (1.01–1.16)  0.87 (0.80–0.94)  
Total cancer (women)PhetTotal cancer (men)PhetBreast cancerPhetProstate cancerPhetColorectal cancerPhetLung cancerPhet
Overall 1.03 (1.01–1.06)  1.03 (1.01–1.06)  1.10 (1.07–1.14)  1.09 (1.05–1.12)  1.07 (1.02–1.11)  0.91 (0.86–0.96)  
Sex          0.23  0.003 
 Male —  1.03 (1.01–1.06)  —  1.09 (1.05–1.12)  1.10 (1.03–1.17)  0.85 (0.79–0.91)  
 Female 1.03 (1.01–1.06)  —  1.10 (1.07–1.14)  —  1.04 (0.98, 1.10)  1.00 (0.92–1.08)  
Menopausal status, among women only  0.07    0.06    0.28  0.29 
 Premenopausal 1.08 (1.02–1.14)  —  1.17 (1.08–1.26)  —  1.19 (0.97–1.46)  0.80 (0.54–1.19)  
 Postmenopausal 1.02 (0.99–1.04)  —  1.08 (1.03–1.12)  —  1.06 (0.99–1.14)  0.99 (0.91–1.08)  
BMI, kg/m2  0.30  0.10  0.59  0.25  1.00  0.33 
 <25 1.01 (0.98–1.05)  1.00 (0.96–1.05)  1.12 (1.05–1.18)  1.05 (0.98–1.12)  1.07 (0.98–1.16)  0.88 (0.80–0.97)  
 ≥25 1.04 (1.01–1.07)  1.04 (1.02–1.07)  1.10 (1.05–1.14)  1.10 (1.06–1.14)  1.07 (1.01–1.12)  0.93 (0.87–0.997)  
Smoking status  0.58  0.0003  0.81  0.72  0.89  0.0005 
 Never-smoker 1.04 (1.01–1.07)  1.08 (1.05–1.12)  1.10 (1.05–1.15)  1.08 (1.00–1.15)  1.07 (1.00–1.14)  1.14 (0.99–1.30)  
 Ever-smoker 1.02 (0.99–1.06)  1.00 (0.97–1.03)  1.11 (1.05–1.17)  1.09 (1.05–1.13)  1.06 (1.00–1.13)  0.88 (0.83–0.93)  
Alcohol consumption  0.84  0.23  0.37  0.89  0.87  0.54 
 Less than one drink per week 1.03 (0.99–1.06)  1.01 (0.96–1.05)  1.08 (1.02–1.14)  1.09 (1.03–1.14)  1.07 (0.99–1.16)  0.94 (0.85–1.03)  
 One drink or more per week 1.03 (1.00–1.06)  1.04 (1.01–1.07)  1.12 (1.07–1.17)  1.09 (1.04–1.14)  1.06 (1.01–1.12)  0.90 (0.84–0.97)  
Follow-up duration, years  0.43  0.45  0.56  0.24  0.60  0.82 
 <5 1.02 (1.00–1.05)  1.04 (1.01–1.07)  1.10 (1.05–1.14)  1.07 (1.03–1.12)  1.06 (1.00–1.12)  0.91 (0.85–0.98)  
 ≥5 1.04 (1.00–1.08)  1.02 (0.98–1.06)  1.12 (1.05–1.19)  1.12 (1.06–1.18)  1.08 (1.00–1.17)  0.92 (0.84–1.01)  
Sex hormone binding globulin, nmol/L  0.92  0.40  0.21  0.45  0.29  0.19 
 <Median 1.03 (1.00–1.06)  1.04 (1.01–1.08)  1.11 (1.06–1.17)  1.07 (1.01–1.12)  1.10 (1.03–1.17)  0.95 (0.87–1.04)  
 ≥Median 1.03 (0.99–1.06)  1.02 (0.99–1.05)  1.06 (1.01–1.12)  1.09 (1.04–1.15)  1.04 (0.98–1.11)  0.88 (0.82–0.95)  
Testosterone, nmol/L  0.47  0.66  0.72  0.53  0.47  0.26 
 <Median 1.02 (0.98–1.06)  1.04 (1.01–1.07)  1.08 (1.02–1.14)  1.08 (1.03–1.13)  1.05 (0.98–1.11)  0.92 (0.85–1.00)  
 ≥Median 1.04 (1.01–1.07)  1.03 (1.00–1.06)  1.09 (1.04–1.15)  1.10 (1.05–1.15)  1.08 (1.01–1.16)  0.87 (0.80–0.94)  

Note: Adjusted for age (5-year categories), assessment center, first two principal components, sex (for cancers in men and women), alcohol consumption (never, special occasions only, 1–3 times per month, 1–2 times per week, 3–4 times per week, daily/almost daily), smoking status (never, former, current), physical activity on the International Physical Activity Questionnaire (IPAQ) scale (low, moderate, high, missing), height in cm (women: <155, 155–159, 160–164, 165–169, ≥170; men: <165, 165–169, 170–174, 175–179, ≥180), BMI in kg/m2 (<21, 21–24.9, 25–29.9, 30–34.9, ≥35), waist-to-hip ratio (women: ≤0.80, 0.81–0.85, >0.85; men: ≤0.90, 0.91–0.95, >0.95), and average household income (less than £18,000, £18,000 to £30,999, £31,000 to £51,999, £52,000 to £100,000, greater than £100,000, refused, do not know). For female-specific cancers, we additionally adjusted for menopausal status (premenopausal, postmenopausal, unknown menopausal status), age at menarche (<12, 12–13, ≥14, missing), parity (nulliparous, 1, 2, 3, ≥4), age at first live birth (nulliparous, <20, 20–24, 25–29, ≥30), ever used oral contraceptives (yes, no), and ever used hormone replacement therapy (yes, no). For breast, prostate, colorectal, and lung cancer, we additionally adjusted for family history of the respective cancer in a first-degree relative (yes, no). For breast cancer, we additionally adjusted for prior history of breast cancer screening/mammograms (ever, never). Bold text indicates statistically significant heterogeneity.

To account for potential subclinical liver disease/injury that may confound the relationship between IGF-1 and liver cancer, we additionally adjusted for prediagnostic ALT, AST, GGT, and total bilirubin. The associations were not materially altered, HR (95% CI): 0.45 (0.37, 0.55; Supplementary Table S3). For kidney cancer, we additionally adjusted for baseline creatinine and cystatin-C, to account for baseline kidney function, and observed similar results, 1.08 (0.98, 1.18; Supplementary Table S6).

Our study demonstrated divergent associations between IGF-1 and site-specific cancers, with increased risks for breast, prostate, colorectal, kidney, thyroid cancers, and melanoma, and lower risks for lung, ovarian, HNC, and liver cancers. Our study also shows that IGF-1 has a modest positive association with total cancer risk (excluding nonmelanoma skin cancer) in both men and women. To our knowledge, our study represents the first large-scale prospective investigation of circulating IGF-1 and incident overall cancer.

The positive association between IGF-1 and breast cancer (HR = 1.24) is consistent with a prior pooled analysis (2), which found an OR of 1.28 (95% CI, 1.14–1.44) comparing extreme quintiles of IGF-1. Associations were comparable for premenopausal and postmenopausal women in our study, which is consistent with prior evidence (2). In addition, we demonstrated similar associations for low versus high SHBG or testosterone, suggesting IGF-1 may elevate breast cancer risk independently of other endogenous hormones. During the preparation of our manuscript, an independent analysis of the UK Biobank with a smaller sample of incident breast cancer cases (N = 4,360) obtained nearly identical findings (23). Moreover, the Mendelian randomization analyses conducted in this study showed a positive association between IGF-1 and overall and ER+ breast cancer, further supporting IGF-1 as a potential causal factor. Our finding of a positive association between IGF-1 and prostate cancer is consistent with a prior individual participant-level meta-analysis, with OR (95% CI) comparing extreme quintiles: 1.29 (1.16–1.43; ref. 24). Similarly, we confirmed prior findings of a positive association between IGF-1 and colorectal cancer, RR (95% CI) per 1-SD increase in IGF1: 1.07 (1.01–1.14; ref. 7). Finally, our results for thyroid cancer supports a European Prospective Investigation into Cancer report, where higher IGF-1 was positively associated with differentiated thyroid carcinoma, OR (95% CI) per doubling of IGF-1: 1.48 (1.06–2.08; ref. 25).

Prior research has suggested that IGF-1 may play a role in the incidence and progression of renal cell carcinoma (26). We found a significant positive association between IGF-1 and kidney cancer that remained robust to further adjustment for baseline kidney function. Our finding stood in contrast to a nested case–control study of male smokers (100 incident cases), which found an inverse association between IGF-1 and kidney cancer (27). In addition, our finding of a positive association between IGF-1 and melanoma risk stood in contrast with existing studies. One case–control study with IGF-1 measured after melanoma diagnosis found an inverse association (28). Another study utilized a nested case–control design and did not show a significant association between IGF-1 and melanoma (29). Hence, future prospective investigations are warranted on IGF-1 and incident kidney cancer or melanoma.

For lung cancer, a prior meta-analysis of six nested case–control studies (1,043 cases) found no significant associations with IGF-1, OR (95% CI) for high versus low IGF-1: 1.05 (0.80–1.37; ref. 8). Our study showed a modest inverse association between IGF-1 and lung cancer, which was confined to ever-smokers and men. Notably, smoking may impact IGF-1, as individuals who smoke tend to have lower circulating IGF-1 than those who abstain (30). In the UK Biobank, we observed that IGF-1 in never-smokers was 0.61 nmol/L higher on average than in ever-smokers, with an even larger difference between nonsmokers and smokers among men than among women (1.00 vs. 0.43 nmol/L, P < 0.0001). This difference may be partially accounted for by greater smoking intensity among men (5.4 higher pack-years). In turn, residual confounding from smoking is possible as reporting pack-years of smoking may differ between men and women. Alternatively, these interactions may be different depending on lung cancer subtypes, because smoking is a stronger risk factor for small cell and squamous cell lung cancer compared with adenocarcinomas (31). On the other hand, we observed a borderline positive association between IGF-1 and lung cancer risk among never-smokers. Similarly, residual confounding by smoking may have led to our finding of a modest inverse association between IGF-1 and HNC, particularly because studies have suggested IGF-1 receptor as a target for HNC (32). Indeed, in a post hoc analysis, we found that there was a trend toward increased risk for HNC among never-smokers: 1.07 (0.91–1.26), whereas there was a protective association among ever-smokers: 0.83 (0.74–0.93), Pint = 0.01. Nevertheless, we are not aware of other prospective studies that have assessed the association between IGF-1 and HNC. Hence, more research is needed to elucidate whether IGF-1 could play a role in lung or head and neck carcinogenesis, and how smoking may modify this relationship.

Similar to our observations, higher IGF-1 has been found to be associated with a lower risk of ovarian cancer in one prior meta-analysis (9). The included studies tended to observe a stronger inverse association between IGF-1 and ovarian cancer in younger or premenopausal women compared with older women. We similarly observed a nominally stronger inverse association in women younger than 55 years, HR (95% CI): 0.68 (0.55–0.85), than those older than 55 years, 0.92 (0.82–1.05), Pint = 0.02. Potential mechanisms for this inverse association between IGF-1 and ovarian cancer remains unclear, though previous studies have suggested that IGF-1 may downregulate the bioactivity of circulating estrogens due to its affinity for both IGF-1 receptors and estrogen receptors (33, 34). Further studies are warranted to elucidate the role that IGF-1 plays in the pathogenesis of ovarian cancer.

The strong inverse association we observed between higher IGF-1 levels and liver cancer is an outlier of all cancers examined, but this finding is consistent with prior studies (35, 36). Epidemiologic and experimental studies have implicated low IGF-1 as a risk factor for the development and progression of nonalcoholic fatty liver disease (37), which is a common risk factor for liver cancer in high-income countries. Moreover, the majority of circulating IGF-1 is produced by hepatocytes in response to growth hormone (GH), and thus lower circulating IGF-1 may reflect hepatic dysfunction (38). Despite adjustments for hepatic markers, we cannot rule out the possibility that undiagnosed liver disease led to lower IGF-1. Additional studies are needed to examine whether subclinical liver damage may perturb the IGF-pathway and whether low IGF-1 is causally related to hepatocellular carcinoma or simply a marker of hepatic injury/dysfunction.

There are several potential mechanisms for the positive association between IGF-1 and cancers. IGF-1 is positively associated with height, and greater early life height velocity and final attained height are both correlated with higher IGF-1 exposures (39). Height has been found in observational and Mendelian randomization studies to be positively associated with multiple malignancies (40–42). Patients who have acromegaly, a condition of excessive GH secretion and in turn higher IGF-1 levels, have an increased risk of colorectal cancer and possible increased risks of breast and prostate cancer (43). Conversely, Laron syndrome patients, who have a resistance to GH stimulation, experience a lower risk of cancer (44). Genetic variants and mutations in the IGF-1 receptor have been found to be associated with increased risk of breast, colorectal, and non–small cell lung cancers (45–47). Moreover, interactions between insulin and the IGF proteins are likely given the structural similarities of these proteins and their receptors. Previous studies have shown that, irrespective of diabetes status, higher circulating insulin and C-peptides were associated with increased risks of breast and colorectal cancer (48). Dysregulated signaling through these two hormones can activate downstream signaling through the MAPK and PI3K pathways, leading to enhanced protein synthesis, cell growth and division, and evasion of apoptosis (49). Therefore, IGF-1 may be a plausible target for the primary prevention of certain cancers. The potential mechanisms for the inverse associations between IGF-1 and cancers of the lung and ovaries are not clear but probably differ from those discussed above. For lung cancer, the borderline positive association among never-smokers is perhaps in line with the mechanisms described above and is likely to be free from residual confounding by smoking. Nevertheless, mechanistic studies and clinical interventions are needed to assess whether and why IGF-1 may selectively elevate the risks of certain cancers while playing a protective role in others.

Our study has several key strengths. The use of a prospective design with large sample size reduces the likelihood of reverse causation and allows for the comprehensive assessment of the relationship between circulating IGF-1 and multiple common and rare cancers. Our results were robust to extensive statistical adjustments and across a wide variety of participant characteristics. The measurement of IGF-1 using a common assay at a central laboratory can help reduce laboratory variations and random errors that could bias or attenuate associations (17). Several limitations warrant mentioning. Information on cancer stage, grade, subtype, and morphology were not available, so we could not examine whether associations with IGF-1 differed by these characteristics. For cancers with lower incidence rates, we may have lacked statistical power to detect an association if one truly exists. False positive findings due to the examination of multiple cancers is possible, though associations observed for most cancers remained significant even after correcting for multiple testing. We could not assess associations with circulating IGF-binding proteins, which could modify the associations for IGF-1. The median follow-up duration for participants in our study was around 7.2 years, which may be relatively short for several cancer types. While we observed consistent associations according to duration of follow-up, future studies with longer follow-up are warranted. Because of the observational nature of this study, we cannot rule out unmeasured confounding. Mendelian randomization studies and RCTs targeting the suppression of IGF-1 will be necessary to determine whether IGF-1 is causally associated with specific cancers.

In conclusion, our results suggest that serum IGF-1 may be a risk factor for specific invasive cancers. Whether our results indicate a causal role for IGF-1 and whether targeting IGF-1 has therapeutic benefits for the primary prevention of these cancers warrant further research.

No potential conflicts of interest were disclosed.

The funders had no role in the design, implementation, data management/analysis, interpretation of the study, or the decision to submit for publication.

F. Qian: Conceptualization, formal analysis, investigation, visualization, writing–original draft, writing–review and editing. D. Huo: Conceptualization, data curation, software, formal analysis, supervision, funding acquisition, validation, investigation, methodology, writing–original draft, project administration, writing–review and editing.

The current analyses were approved under the application number 49564. The authors thank the participants, investigators, and staff of the UK Biobank for providing them with the resources to pursue this research.

F. Qian was supported by the John D. Arnold Scientific Research Prize from the University of Chicago Pritzker School of Medicine. This study was partially supported by the Breast Cancer Research Foundation and NCI (R01 CA242929; to D. Huo).

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.
Le Roith
D
. 
Seminars in medicine of the Beth Israel Deaconess Medical Center. Insulin-like growth factors
.
N Engl J Med
1997
;
336
:
633
40
.
2.
Endogenous
H
,
Breast Cancer Collaborative G
,
Key
TJ
,
Appleby
PN
,
Reeves
GK
,
Roddam
AW
. 
Insulin-like growth factor 1 (IGF1), IGF binding protein 3 (IGFBP3), and breast cancer risk: pooled individual data analysis of 17 prospective studies
.
Lancet Oncol
2010
;
11
:
530
42
.
3.
Kaaks
R
,
Johnson
T
,
Tikk
K
,
Sookthai
D
,
Tjonneland
A
,
Roswall
N
, et al
Insulin-like growth factor I and risk of breast cancer by age and hormone receptor status-a prospective study within the EPIC cohort
.
Int J Cancer
2014
;
134
:
2683
90
.
4.
Christopoulos
PF
,
Msaouel
P
,
Koutsilieris
M
. 
The role of the insulin-like growth factor-1 system in breast cancer
.
Mol Cancer
2015
;
14
:
43
.
5.
Chan
JM
,
Stampfer
MJ
,
Giovannucci
E
,
Gann
PH
,
Ma
J
,
Wilkinson
P
, et al
Plasma insulin-like growth factor-I and prostate cancer risk: a prospective study
.
Science
1998
;
279
:
563
6
.
6.
Shi
R
,
Berkel
HJ
,
Yu
H
. 
Insulin-like growth factor-I and prostate cancer: a meta-analysis
.
Br J Cancer
2001
;
85
:
991
6
.
7.
Rinaldi
S
,
Cleveland
R
,
Norat
T
,
Biessy
C
,
Rohrmann
S
,
Linseisen
J
, et al
Serum levels ofIGF-I, IGFBP-3 and colorectal cancer risk: results from the EPIC cohort, plus a meta-analysis of prospective studies
.
Int J Cancer
2010
;
126
:
1702
15
.
8.
Cao
H
,
Wang
G
,
Meng
L
,
Shen
H
,
Feng
Z
,
Liu
Q
, et al
Association between circulating levels of IGF-1 and IGFBP-3 and lung cancer risk: a meta-analysis
.
PLoS One
2012
;
7
:
e49884
.
9.
Li
Y
,
Li
Y
,
Zhang
J
,
Zheng
C
,
Zhu
H
,
Yu
H
, et al
Circulating insulin-like growth factor-1 level and ovarian cancer risk
.
Cell Physiol Biochem
2016
;
38
:
589
97
.
10.
Rohrmann
S
,
Linseisen
J
,
Becker
S
,
Allen
N
,
Schlehofer
B
,
Overvad
K
, et al
Concentrations of IGF-I and IGFBP-3 and brain tumor risk in the European Prospective Investigation into Cancer and Nutrition
.
Cancer Epidemiol Biomarkers Prev
2011
;
20
:
2174
82
.
11.
Kim
HJ
,
Kwon
H
,
Lee
JW
,
Kim
HJ
,
Lee
SB
,
Park
HS
, et al
Metformin increases survival in hormone receptor-positive, HER2-positive breast cancer patients with diabetes
.
Breast Cancer Res
2015
;
17
:
64
.
12.
Lega
IC
,
Austin
PC
,
Gruneir
A
,
Goodwin
PJ
,
Rochon
PA
,
Lipscombe
LL
. 
Association between metformin therapy and mortality after breast cancer: a population-based study
.
Diabetes Care
2013
;
36
:
3018
26
.
13.
Arrieta
O
,
Barron
F
,
Padilla
MS
,
Aviles-Salas
A
,
Ramirez-Tirado
LA
,
Arguelles Jimenez
MJ
, et al
Effect of metformin plus tyrosine kinase inhibitors compared with tyrosine kinase inhibitors alone in patients with epidermal growth factor receptor-mutated lung adenocarcinoma: a phase 2 randomized clinical trial
.
JAMA Oncol
2019
;
5
:
e192553
.
14.
Allen
NE
,
Sudlow
C
,
Peakman
T
,
Collins
R
,
Biobank
UK
. 
UK Biobank data: come and get it
.
Sci Transl Med
2014
;
6
:
224ed4
.
15.
Elliott
P
,
Peakman
TC
,
Biobank
UK
. 
The UK Biobank sample handling and storage protocol for the collection, processing and archiving of human blood and urine
.
Int J Epidemiol
2008
;
37
:
234
44
.
16.
Hosgood
HD
,
Gunter
MJ
,
Murphy
N
,
Rohan
TE
,
Strickler
HD
. 
The relation of obesity-related hormonal and cytokine levels with multiple myeloma and non-Hodgkin lymphoma
.
Front Oncol
2018
;
8
:
103
.
17.
UK Biobank
.
UK Biobank Biomarker Project - companion document to accompany serum biomarker data. Version 1.0
.
Cardiff (UK)
:
UK Biobank
; 
2019 Nov 3
.
Available from
: https://biobank.ndph.ox.ac.uk/showcase/showcase/docs/serum_biochemistry.pdf.
18.
Al-Ajmi
K
,
Lophatananon
A
,
Ollier
W
,
Muir
KR
. 
Risk of breast cancer in the UK Biobank female cohort and its relationship to anthropometric and reproductive factors
.
PLoS One
2018
;
13
:
e0201097
.
19.
Clarke
R
,
Shipley
M
,
Lewington
S
,
Youngman
L
,
Collins
R
,
Marmot
M
, et al
Underestimation of risk associations due to regression dilution in long-term follow-up of prospective studies
.
Am J Epidemiol
1999
;
150
:
341
53
.
20.
MacMahon
S
,
Peto
R
,
Cutler
J
,
Collins
R
,
Sorlie
P
,
Neaton
J
, et al
Blood pressure, stroke, and coronary heart disease. Part 1, prolonged differences in blood pressure: prospective observational studies corrected for the regression dilution bias
.
Lancet
1990
;
335
:
765
74
.
21.
Fine
JP
,
Gray
RJ
. 
A proportional hazards model for the subdistribution of a competing risk
.
J Am Statist Assoc
1999
;
94
:
496
509
.
22.
Vandenbroucke
JP
,
von Elm
E
,
Altman
DG
,
Gotzsche
PC
,
Mulrow
CD
,
Pocock
SJ
, et al
Strengthening the Reporting of Observational Studies in Epidemiology (STROBE): explanation and elaboration
.
Epidemiology
2007
;
18
:
805
35
.
23.
Murphy
N
,
Knuppel
A
,
Papadimitriou
N
,
Martin
RM
,
Tsilidis
KK
,
Smith-Byrne
K
, et al
Insulin-like growth factor-1, insulin-like growth factor-binding protein-3, and breast cancer risk: observational and Mendelian randomization analyses with approximately 430 000 women
.
Ann Oncol
2020
;
31
:
641
9
.
24.
Travis
RC
,
Appleby
PN
,
Martin
RM
,
Holly
JMP
,
Albanes
D
,
Black
A
, et al
A meta-analysis of individual participant data reveals an association between circulating levels of IGF-I and prostate cancer risk
.
Cancer Res
2016
;
76
:
2288
300
.
25.
Schmidt
JA
,
Allen
NE
,
Almquist
M
,
Franceschi
S
,
Rinaldi
S
,
Tipper
SJ
, et al
Insulin-like growth factor-i and risk of differentiated thyroid carcinoma in the European Prospective Investigation into Cancer and Nutrition
.
Cancer Epidemiol Biomarkers Prev
2014
;
23
:
976
85
.
26.
Tracz
AF
,
Szczylik
C
,
Porta
C
,
Czarnecka
AM
. 
Insulin-like growth factor-1 signaling in renal cell carcinoma
.
BMC Cancer
2016
;
16
:
453
.
27.
Major
JM
,
Pollak
MN
,
Snyder
K
,
Virtamo
J
,
Albanes
D
. 
Insulin-like growth factors and risk of kidney cancer in men
.
Br J Cancer
2010
;
103
:
132
5
.
28.
Park
SL
,
Setiawan
VW
,
Kanetsky
PA
,
Zhang
ZF
,
Wilkens
LR
,
Kolonel
LN
, et al
Serum insulin-like growth factor-I and insulin-like growth factor binding protein-3 levels with risk of malignant melanoma
.
Cancer Causes Control
2011
;
22
:
1267
75
.
29.
Bradbury
KE
,
Appleby
PN
,
Tipper
SJ
,
Travis
RC
,
Allen
NE
,
Kvaskoff
M
, et al
Circulating insulin-like growth factor I in relation to melanoma risk in the European Prospective Investigation into Cancer and Nutrition
.
Int J Cancer
2019
;
144
:
957
66
.
30.
Gapstur
SM
,
Kopp
P
,
Chiu
BC
,
Gann
PH
,
Colangelo
LA
,
Liu
K
. 
Longitudinal associations of age, anthropometric and lifestyle factors with serum total insulin-like growth factor-I and IGF binding protein-3 levels in Black and White men: the CARDIA Male Hormone Study
.
Cancer Epidemiol Biomarkers Prev
2004
;
13
:
2208
16
.
31.
Pesch
B
,
Kendzia
B
,
Gustavsson
P
,
Jockel
KH
,
Johnen
G
,
Pohlabeln
H
, et al
Cigarette smoking and lung cancer–relative risk estimates for the major histological types from a pooled analysis of case-control studies
.
Int J Cancer
2012
;
131
:
1210
9
.
32.
Lehman
CE
,
Khalil
AA
,
Axelrod
MJ
,
Dougherty
MI
,
Schoeff
SS
,
Taniguchi
LE
, et al
Antitumor effect of insulin-like growth factor-1 receptor inhibition in head and neck squamous cell carcinoma
.
Laryngoscope
2020
;
130
:
1470
8
.
33.
Lukanova
A
,
Kaaks
R
. 
Endogenous hormones and ovarian cancer: epidemiology and current hypotheses
.
Cancer Epidemiol Biomarkers Prev
2005
;
14
:
98
107
.
34.
Pollak
M
. 
Insulin and insulin-like growth factor signalling in neoplasia
.
Nat Rev Cancer
2008
;
8
:
915
28
.
35.
Major
JM
,
Stolzenberg-Solomon
RZ
,
Pollak
MN
,
Snyder
K
,
Virtamo
J
,
Albanes
D
. 
Insulin-like growth factors and liver cancer risk in male smokers
.
Br J Cancer
2010
;
103
:
1089
92
.
36.
Wang
J
,
Li
YC
,
Deng
M
,
Jiang
HY
,
Guo
LH
,
Zhou
WJ
, et al
Serum insulin-like growth factor-1 and its binding protein 3 as prognostic factors for the incidence, progression, and outcome of hepatocellular carcinoma: a systematic review and meta-analysis
.
Oncotarget
2017
;
8
:
81098
108
.
37.
Takahashi
Y
. 
The role of growth hormone and insulin-like growth factor-I in the liver
.
Int J Mol Sci
2017
;
18
:
1447
.
38.
Olivecrona
H
,
Hilding
A
,
Ekstrom
C
,
Barle
H
,
Nyberg
B
,
Moller
C
, et al
Acute and short-term effects of growth hormone on insulin-like growth factors and their binding proteins: serum levels and hepatic messenger ribonucleic acid responses in humans
.
J Clin Endocrinol Metab
1999
;
84
:
553
60
.
39.
Stefan
N
,
Haring
HU
,
Hu
FB
,
Schulze
MB
. 
Divergent associations of height with cardiometabolic disease and cancer: epidemiology, pathophysiology, and global implications
.
Lancet Diabetes Endocrinol
2016
;
4
:
457
67
.
40.
Zhang
B
,
Shu
XO
,
Delahanty
RJ
,
Zeng
C
,
Michailidou
K
,
Bolla
MK
, et al
Height and breast cancer risk: evidence from prospective studies and Mendelian randomization
.
J Natl Cancer Inst
2015
;
107
:
djv219
.
41.
Qian
F
,
Wang
S
,
Mitchell
J
,
McGuffog
L
,
Barrowdale
D
,
Leslie
G
, et al
Height and body mass index as modifiers of breast cancer risk in BRCA1/2 mutation carriers: a Mendelian randomization study
.
J Natl Cancer Inst
2019
;
111
:
350
64
.
42.
Khankari
NK
,
Shu
XO
,
Wen
W
,
Kraft
P
,
Lindstrom
S
,
Peters
U
, et al
Association between adult height and risk of colorectal, lung, and prostate cancer: results from meta-analyses of prospective studies and Mendelian randomization analyses
.
PLoS Med
2016
;
13
:
e1002118
.
43.
Jenkins
PJ
. 
Acromegaly and cancer
.
Horm Res
2004
;
62
:
108
15
.
44.
Janecka
A
,
Kolodziej-Rzepa
M
,
Biesaga
B
. 
Clinical and molecular features of laron syndrome, a genetic disorder protecting from cancer
.
In Vivo
2016
;
30
:
375
81
.
45.
Neuhausen
SL
,
Brummel
S
,
Ding
YC
,
Singer
CF
,
Pfeiler
G
,
Lynch
HT
, et al
Genetic variation in insulin-like growth factor signaling genes and breast cancer risk among BRCA1 and BRCA2 carriers
.
Breast Cancer Res
2009
;
11
:
R76
.
46.
Reinmuth
N
,
Kloos
S
,
Warth
A
,
Risch
A
,
Muley
T
,
Hoffmann
H
, et al
Insulin-like growth factor 1 pathway mutations and protein expression in resected non-small cell lung cancer
.
Hum Pathol
2014
;
45
:
1162
8
.
47.
Winder
T
,
Zhang
W
,
Yang
D
,
Ning
Y
,
Bohanes
P
,
Gerger
A
, et al
Germline polymorphisms in genes involved in the IGF1 pathway predict efficacy of cetuximab in wild-type KRAS mCRC patients
.
Clin Cancer Res
2010
;
16
:
5591
602
.
48.
Gallagher
EJ
,
LeRoith
D
. 
The proliferating role of insulin and insulin-like growth factors in cancer
.
Trends Endocrinol Metab
2010
;
21
:
610
8
.
49.
Hakuno
F
,
Takahashi
SI
. 
IGF1 receptor signaling pathways
.
J Mol Endocrinol
2018
;
61
:
T69
86
.