Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Oct 1.
Published in final edited form as: Nat Rev Clin Oncol. 2020 Nov 20;18(4):230–243. doi: 10.1038/s41571-020-00445-1

Rising incidence of early-onset colorectal cancer: a call for action

Naohiko Akimoto 1,2, Tomotaka Ugai 1,3, Rong Zhong 1,4,5, Tsuyoshi Hamada 6, Kenji Fujiyoshi 1,7, Marios Giannakis 8,9,10, Kana Wu 3,4,11, Yin Cao 12,13,14, Kimmie Ng 8, Shuji Ogino 1,3,9,15
PMCID: PMC7994182  NIHMSID: NIHMS1629340  PMID: 33219329

Abstract

Early-onset colorectal cancer (CRC), which occurs in individuals under 50 years of age, has been increasing worldwide, especially in high-income countries, for unknown reasons. Plausible hypotheses for this rise include exposure to potential risk factors such as western-style diet, obesity, physical inactivity, and increased antibiotic use, especially in early life from prenatal to adolescent periods. These exposures may not only cause genetic and epigenetic alterations in colorectal epithelial cells, but also affect the gut microbiota and host immunity. When compared to later-onset CRCs, early-onset CRCs exhibit differential clinical, pathological, and molecular features. While certain existing resources can be utilized to elucidate the etiology of early-onset CRC and inform the development of effective prevention, early detection, and therapeutic strategies, there is a need for additional life-course cohort studies spanning the periods of early-life to young adulthood integrated with prospective biospecimen collections, omics biomarker analyses, and the molecular pathological epidemiology approach.

Introduction

Colorectal cancer (CRC) most often affects elderly individuals1. While overall CRC incidence rates have remained stable or declined in many high-income countries, incidence of early-onset CRC (generally defined as CRC diagnosed before the age of 50 years) has recently been increasing worldwide2,3. In this article, contrasting with “early-onset CRC”, we use the term “later-onset CRC” in patients aged 50 years or above. The reasons underlying this rise of early-onset CRC are poorly understood. This article discusses the current evidence for salient characteristics of early-onset CRC and its potential etiological mechanisms. A better understanding of the characteristics and etiology of early-onset CRC can promote effective prevention, early detection, and treatment strategies.

While we discuss differences in epidemiological, clinical, pathological, and molecular features between early-onset and later-onset CRC, there may not be a sharp dichotomy of the features in CRC patients aged 49 vs. those aged 50. Those characteristics of CRC may change in any way, with increasing age at diagnosis as well as changes in other key variables such as tumor location. Accordingly, we need to consider heterogeneity within early-onset CRC patients and within later-onset CRC patients.

What is already known?

Epidemiology

CRC is the third most common cancer worldwide with 1.8 million new cases and the second most common cause of cancer death with over 880,000 deaths in 20184. CRC incidence varies substantially across continents5,6 with the highest in Australia/New Zealand and the lowest in Africa and South-Central Asia4. These differences may be attributable to variations in genetic susceptibility, socioeconomic status, environmental exposures, diets, other lifestyle factors, and/or screening practices7,8. For the past two decades, CRC incidence has remained stable or declined in high-income countries, while low/middle-income countries with historically lower CRC rates have documented an increase in CRC incidence5. Established CRC risk factors include (but are not limited to) red and processed meat, alcohol, obesity, inflammatory bowel disease (IBD), CRC family history, and CRC-predisposing genetic variants, whereas protective factors include aspirin use, high systemic vitamin D level, high folate intake, and physical activity8.

In recent decades, early-onset CRC incidence has been increasing in both men and women across many countries2,9-11. The average annual percent changes in early-onset CRC incidence were 4.0% in New Zealand, 2.8% in Canada and Australia, and 2.2% in the U.S. during 2008-20122. In the U.S., early-onset CRC incidence has been increasing since the mid 1990s12, and the age-adjusted early-onset CRC incidence per 100,000 persons was 5.9 cases in 2000 and 8.4 cases in 2017. Increases in early-onset CRC have also been documented in most European countries. Early-onset CRC incidence (per 100,000 persons) increased from 0.8 to 2.3 cases in individuals aged 20-29 years during1990-2016, from 2.8 to 6.4 cases in those aged 30-39 during 2006-2016, and from 15.5 to 19.2 cases in those aged 40-49 during 2005-2016. The average annual percent changes in early-onset CRC incidence were 7.9% in individuals aged 20-29, 4.9% in those aged 30-39, and 1.6% in those aged 40-49 during 2004-20163. Taken together, early-onset CRC now represents a significant cancer burden among younger adults.

The increase of early-onset CRC incidence in the U.S. was initially largely driven by rectal cancer12. The annual percent change of rectal cancer incidence was 3.2% for ages 20-39 during 1980-2013, and 2.3% for ages 40-49 during 1991-2013, while that of colon cancer incidence was 2.4% for ages 20-29 during 1983-2013, 1.0% for ages 30-39 during 1988-2013, and 1.3% for ages 40-49 during 1996-201312. Since 2012 early-onset CRC incidence has increased similarly for colon and rectum with the annual percent change of approximately 1.8%1. The rise in early-onset CRC incidence appeared more prominent for colon cancer than for rectal cancer in Europe3.

The upward trend of early-onset CRC incidence in most western countries since the 1980s3,12-15 seems to reflect a birth cohort effect, which means that temporal changes in certain risk factors might have differentially affected each age-group (i.e., birth cohort) with increased risk being carried forward to later time. There is a possibility that the observed birth cohort effect might be caused by secular trends in risk factor exposures during early life, typically referring to prenatal to adolescent life (in contrast to adulthood), and their delayed effects on CRC incidence12,16. Considering that the rise in CRC incidence has generally followed the westernization of lifestyle, early-onset CRC will likely become an emerging issue in regions where western lifestyle had not been widely adopted until recently. Indeed, increasing incidence of early-onset CRC has already been documented in Taiwan, Korea, Japan, and Hong Kong17. Moreover, incidence of early-onset cancers of other body sites that share risk factors with CRC may also increase18,19.

Most epidemiological investigations that examined potential risk factors for early-onset CRC were case-control studies20-22, while relatively few studies prospectively examined exposure data collected before CRC diagnosis23,24 (Table 1). Potential early-onset CRC risk factors shown in the case-control studies include male sex21, race (black and Asian ethnicities)21, family history of CRC20,21, alcohol20, weight loss of ≥5 kg (within five-year period preceding colonoscopy)22, processed meat intake20, and inflammatory bowel disease (IBD)21. On the contrary, aspirin use22 and higher intake of vegetables20, citrus fruits20, fish20, β-carotene20, vitamin C20, vitamin E20, and folate20 have been associated with lower risk of early-onset CRC20,22. In the two published prospective studies, sedentary lifestyle (measured as TV watching time)23 and obesity24 were associated with higher incidence of early-onset CRC. However, the association between obesity and early-onset CRC remains inconclusive with some studies showing positive associations18,24 and another showing opposite results22.

Table 1.

Epidemiological studies of early-onset colorectal cancer and early-onset adenoma.

Ref
No.
Authors
(year)
Country Study design Study population, No. Case, No.
(diagnosed age)
Risk factors, OR/HR (95% CI)*
20 Rosato et al. (2013) Italia and Switzer-land Case-control study 329 EOCRC and 1,361 age-matched controls EOCRC, 329 (≤45) Family history of CRC in first-degree relatives (yes vs no), 4.5 (2.6–7.7); alcohol (≥14 drinks/week vs never drinking), 1.6 (1.1–2.2); processed meat (highest vs lowest tertile), 1.6 (1.1-2.2); vegetables (highest vs lowest tertile), 0.40 (0.28-0.56); citrus fruit (highest vs lowest tertile), 0.61 (0.45-0.84); fish (highest vs lowest tertile), 0.78 (0.60-1.00); β-carotene (highest vs lowest tertile), 0.52 (0.37-0.72); vitamin C (highest vs lowest tertile), 0.68 (0.49-0.94); vitamin E (highest vs lowest tertile), 0.38 (0.26-0.58); folate (highest vs lowest tertile), 0.59 (0.40-0.86).
123 Kim et al. (2016) Korea Cross-sectional study 564 advanced adenoma and CRC and 59,782 controls Kangbuk Samsung Health Study Advanced adenoma and EOCRC, 564 (<50) Age (per year), 1.08 (1.07-1.10); male (vs female), 1.3 (1.0-1.6); smoking (current vs never/former): 1.4 (1.2-1.6); CRC family history (yes vs no), 1.5 (1.0-2.1); diabetes mellitus-related factors (yes vs no), 1.3 (1.1-1.5); obesity (yes vs no), 1.2 (1.0-1.5); low-density lipoprotein-cholesterol (per mg/dL), 1.01 (1.01-1.02); carcinoembryonic antigen (per ng/mL), 1.04 (1.01-1.09).
23 Nguyen et al. (2018) United States Prospective cohort study 89,278 females The Nurses’ Health Study II EOCRC, 118 Sedentary TV viewing time (>14 vs ≤7 hours per week), 1.69 (1.07-2.67).
21 Gausman et al. (2019) United States Case-control study 269 EOCRC, 2,802 LOCRC, and 1,122 controls (age-matched, cancer-free individuals) EOCRC, 269; LOCRC, 2802 EOCRC vs LOCRC: male (vs female), 1.4 (1.1-1.9); non-Hispanic black (vs non-Hispanic white), 1.7 (1.1-2.7); non-Hispanic Asian (vs non-Hispanic white), 2.6 (1.6-4.2); IBD (yes vs no), 3.0 (1.2-6.6); family history of CRC (yes vs no), 2.9 (1.9-4.3).
EOCRC vs controls: male (vs female), 1.9 (1.4-2.5); family history of CRC (yes vs no), 8.6 (4.8-15).
160 Kim et al. (2019) Korea Cross-sectional study 622 advanced adenoma and CRC and 71,734 controls Kangbuk Samsung Health Study Advanced adenoma and EOCRC, 622 (20-39) Diagnosed 20-29 years old: obesity (BMI ≥25 vs <25 kg/m2), 2.5 (1.2-4.9); waist circumference (>90 cm in men or >80 cm in women vs ≤90 cm in men or ≤80 cm in women), 2.3 (1.1-4.5); elevated triglyceride level (≥150 vs <150 mg/dL), 3.0 (1.4-6.2).
Diagnosed 30-39 years old: age (per year), 1.09 (1.06-1.13); smoking (current/former vs never), 1.3 (1.1-1.6); alcohol intake (≥20 vs <20 g/day), 1.3 (1.1-1.6); BMI (≥25 vs <25 kg/m2), 1.3 (1.1-1.6); waist circumference (>90 cm in men or >80 cm in women vs ≤90 cm in men or ≤80 cm in women), 1.3 (1.1-1.6).
24 Liu et al. (2019) United States Prospective cohort study 85,256 females the Nurses’ Health Study II EOCRC, 114 Obese (BMI ≥30 vs 19-23 kg/m2), 2.0 (1.2-3.3); BMI (≥23 vs 18.5-21 kg/m2) at 18 years of age, 1.6 (1.0-2.6); weight gain since 18 years of age (≥40 kg vs loss or gain <5 kg), 2.2 (1.0-4.6).
22 Low et al. (2020) United States Case-control study 651 EOCRC and 67,416 controls EOCRC, 651 Age (per year), 1.05 (1.03-1.07); male (vs female), 2.2 (1.7-2.9); aspirin use (yes vs no), 0.7 (0.5-0.8); overweight (vs normal weight), 0.69 (0.55-0.87); obese (vs normal weight), 0.69 (0.55-0.86); weight loss ≥5 kg within 5-year period preceding colonoscopy (yes vs no), 2.2 (1.8-2.8).

Studies with 200 or fewer cases may not be listed except for studies examining key features.

*

Statistically significant using two-sided p <0.05.

Abbreviations: BMI, body mass index; CI, confidence interval; CRC, colorectal cancer; EOCRC, early-onset colorectal cancer; HR, hazard ratio; IBD, inflammatory bowel disease; LOCRC, later-onset colorectal cancer; OR, odds ratio.

Certain socioeconomic factors, such as race/ethnicity, household income, education levels, and rural vs urban residence, have been associated with increased CRC incidence or mortality25. While African Americans have exhibited a relatively high but stable incidence of early-onset CRC, non-Hispanic whites have experienced a sharp increase in early-onset CRC incidence2,26. African American children and adolescents have relatively high prevalence of obesity and type 2 diabetes27,28. Nonetheless, it remains unclear why whites have shown such a rapid increase in early-onset CRC incidence but African Americans have not1,26. In terms of prognosis, African Americans have been associated with higher mortality compared to whites among early-onset CRC patients, and this difference appears to be attenuated among later-onset CRC patients29.

In general, case-control studies sample hospital-based controls or colonoscopy controls, neither of which were random samples from the general population. There are likely differences in background populations that have given rise to cases vs. controls in case-control studies. Furthermore, differential recall between cases and controls remain an intractable problem when exposure data in cases (but not controls) are recalled after CRC diagnosis. Another issue in any study design is a reverse causation, as their underlying illness even before clinical detection may affect exposure data through changes in physical well-being, appetite, weight, etc. To address the reverse causation problem, data on exposures should ideally be collected long (years to decades) before CRC diagnosis.

Clinical features

Early-onset CRCs most commonly occur in the rectum (35-37%), followed by distal colon (25-26%) and proximal colon (22-23%)1,30, while approximately 29%, 27% and 29% of CRCs diagnosed in individuals aged ≥50 years (hereafter referred to as later-onset CRCs) occur in rectum, distal colon, and proximal colon, respectively30. Certain risk factor associations with later-onset CRC and their effect sizes appear to differ by anatomic sites31. For instance, BMI and waist circumference showed stronger associations with colon cancer compared to rectal cancer, and the associations of height and physical inactivity appeared confined to colon cancer31. In contrast, smoking presented a stronger association with rectal cancer in comparison to colon cancer31. However, it remains unclear whether these differences persist for colon and rectal cancer diagnosed under age 50. Considering the steep rise of early-onset rectal cancer compared to early-onset colon cancer incidence, epidemiological analyses stratified by colorectal subsites should be conducted31.

Patients with early-onset CRC were more likely to be diagnosed at advanced stage (stage III-IV) compared to patients with later-onset CRC32-34. Early-onset CRC patients experienced significantly longer time to diagnosis and longer duration of symptoms compared to older patients34-36. Early-onset CRC has been associated with synchronous and metachronous CRC37. In early-onset CRC patients, lower awareness of CRC, lack of screening, an underappreciation of symptoms, and reluctance to seek medical care may contribute to delayed diagnosis and advanced stage at diagnosis38.

Germline genetics

Table 2 summarizes the studies that have investigated the germline genetic and tumor molecular features of early-onset CRC. Approximately 30% of early-onset CRC patients have a family history of at least one first-degree relative with CRC39-41. There are a number of heritable conditions that can manifest as early-onset CRC (reviewed in detail elsewhere39).

Table 2.

Previous studies of germline genetic and somatic molecular characteristics of early-onset colorectal cancer.

Ref
No.
Authors
(year)
Country No. of cases
(diagnosed age)
MSI-high Pathogenic
germline
mutation
Lynch
syndrome*
Notes
37 Liang et al. (2003) China 138 CRC (<40) vs 339 (≥60) 37/126 (30%) NA NA High-frequency MSI (30% vs 6.3%; p <0.001), and synchronous CRC (6% vs 1%; p = 0.007) were observed in EOCRC compared to LOCRC.
61 Baba et al. (2010) United States 196 CRC (<60) vs 673 (≥60) NA NA NA LINE-1 hypomethylation** were more prevalent in patients aged <60 years than patients aged ≥60 years (p = 0.006). [***Frequencies of LINE-1 methylation level <50% in EOCRC (n = 27) and LOCRC (n = 842) were 22% and 11%, respectively (p = 0.08).]
51 Berg et al. (2010) Norway 23 EOCRC vs 17 LOCRC in non-MSI-high NA NA NA Ten genomic loci listed below were identified as more often altered in tumors from early-onset versus late-onset CRC; gains of 2q35, 10q21.3-22.1, 10q22.3 and 19q13.2-13.31 and losses from 1p31.3, 1q21.1, 2q21.2, 4p16.1-q28.3, 10p11.1 and 19p12.
59 Antelo et al. (2012) Argentina 185 EOCRC vs 135 LOCRC NA NA NA EOCRC showed lower mean LINE-1 methylation level (57%) than LOCRC [MSI-high set (n = 46; 67%), non-MSI-high set (n = 89; 65%), both p < 0.0001].
50 Perea et al. (2014) Spain 82 EOCRC vs 97 LOCRC 12 (15%) NA 10 (12%) Early-onset non-MSI-high CRCs showed different tumor locations (p = 0.02) and more family history of cancer (p <0.001) compared to late-onset non-MSI-high CRCs.
41 Mork et al. (2015) United States 193 EOCRC (≤35) 45 (23%) 67 (35%) 23 (12%) Identifiable hereditary cancer syndromes other than Lynch syndrome were as follows; familial adenomatous polyposis (16 patients), constitutional mismatch repair deficiency (2 patients), biallelic MUTYH mutations (2 patients), and Li-Fraumeni syndrome (1 patient).
52 Arriba et al. (2016) Spain 60 EOCRC vs 86 LOCRC NA NA NA Losses at 1p36, 1p12, 1q21, 9p13, 14q11, 16p13, and 16p12 were significantly more frequent in early-onset CRC, whereas gains at 7q11 and 7q22 were more frequent in later-onset CRC.
40 Pearlman et al. (2017) United States 450 EOCRC 48 (11%) 72 (16%) 37 (8.2%) Germline mutations associated hereditary cancer syndrome except Lynch syndrome were found in the following genes; APC (9 patients), ATM (4 patients), BRCA1 (2 patients), BRCA2 (4 patients), CDKN2A (1 patient), CHECK2 (1 patient), MUTYH (9 patients), PALB2 (2 patients), and SMAD4 (1 patient).
39 Stoffel et al. (2018) United States 315 EOCRC 41 (9.5%) 79 (25%) 56 (18%) Germline mutations associated hereditary cancer syndrome except Lynch syndrome were found in the following genes; APC (10 patients), in BRCA1 (1 patient), CHEK2 (1 patient), MUTYH (8 patients), SMAD4 (2 patients), and TP53 (1 patient).
58 Lieu et al. (2019) United States 1,420 EOCRC (<40), 3,248 EOCRC (40-49) vs 13,550 LOCRC NA NA NA TP53 (FDR < 0.01) and CTNNB1 (FDR = 0.01) mutations were more prevalent in EOCRC patient aged <40 years. In the MSI-high cohort, APC (p <0.001, FDR <0.01) and KRAS (p < 0.001, FDR < 0.01) mutations were more prevalent in EOCRC patients aged <40 years, compared to LOCRC patients aged ≥50 years.
49 Willauer et al. (2019) United States 1,162 EOCRC vs 2,583 LOCRC (2 cohorts; mostly stage IV) 68 (4.5%) in one stage IV cohort of 1525 CRCs NA NA EOCRC had fewer BRAF c.1799T>A (p.V600E) mutations (4.8%) and higher MSI-high (7.4%) compared to LOCRC (8.1%, p < 0.0001; and 3.0%, p = 0.04, respectively).
49 Willauer et al. (2019) United States 82 EOCRC vs 544 LOCRC Same as above NA NA EOCRC patients and ≥70-year-old LOCRC patients (n = 260) had similar CMS distributions, showing higher CMS1 (22-23%) and lower CMS2 (43%) and CMS4 (20-22%) compared to LOCRC patients aged 50-69 years (n = 284) (11% CMS1, 50% CMS2, and 27% CMS4).
30 Archambault et al. (2020) United States 5,479 EOCRC vs. 44,544 LOCRC NA NA 37 (6.4%, among 574 cases) Polygenic risk score developed from 95 CRC-associated common genetic risk variants identified by GWAS were significantly associated with EOCRC.

Studies with 100 or fewer cases having tumor tissue data may not be listed, except for studies examining key features.

*

Lynch syndrome was defined by the presence of a germline pathogenic mutation in any of MMR genes (MLH1, MSH2, MSH6, and PMS2).

**

LINE-1 methylation level (described as 0 to 100) was calculated by the average of methylated cytosine after bisulfite conversion among at the four CpG sites. LINE-1 hypomethylation was defined as < 40%.

***

Unpublished data.

Abbreviations: CMS, consensus molecular subtype; CRC, colorectal cancer; EOCRC, early-onset colorectal cancer; FDR, false discovery rate; GWAS, genome-wide association studies; LINE-1, long-interspersed nucleotide element-1; LOCRC, later-onset colorectal cancer; MAPK, mitogen-activated protein kinase; MMR, mismatch repair; MSI, microsatellite instability; NA, not available.

Lynch syndrome, which is caused by a germline mutation in a DNA mismatch repair (MMR) gene (MLH1, MSH2, MSH6, or PMS2), often manifests as early-onset CRC with high-level microsatellite instability (MSI)42. Studies showed that 16-35% of early-onset CRC patients had hereditary cancer-predisposing syndromes, and 34-71% of these were Lynch syndrome cases39-41, while 45-59% of all MSI-high early-onset CRC patients had Lynch syndrome39,41.

Familial adenomatous polyposis (FAP) is a well-known syndrome characterized by numerous colorectal adenomas in affected individuals8. In a typical FAP family, carriers of a germline pathogenic mutation in APC have a nearly 100% risk of developing CRC by age 408. Increased adoption of genetic testing and prophylactic colectomy have effectively decreased CRC incidence among FAP families43.

There exists a spectrum of cancer predisposition from single-gene Mendelian conditions to polygenic influences plus gene-by-environment (GxE) interactions. A study including over 50,000 CRCs and 58,000 controls showed that a polygenic risk score based on 95 common CRC risk variants was more strongly associated with early-onset CRC than later-onset CRC, particularly in the absence of CRC family history30. Compared to those with the lowest quartile, individuals with the highest quartile polygenic risk score had higher risks of early- and later-onset CRC with odds ratios of 3.7 (95% confidence interval: 3.3-4.2) and 2.9 (95% confidence interval: 2.8-3.0), respectively30. These findings warrant further investigation into the cumulative effect of genetic variants and interactions with environmental exposures to clarify the etiology of early-onset CRC, and guide prevention, early detection, and treatment strategies.

Molecular pathology

CRC represents a pathologically and molecularly heterogenous group of diseases, which are influenced by exogenous and endogenous factors including the tumor immune microenvironment44. The continuum of molecular characteristics of CRC according to bowel subsites is compatible with the interactive roles of the microbiota and immunity in colorectal carcinogenesis45-47. Evidence also indicates genetic, pathological, and molecular heterogeneity in early-onset CRC37,39-41,48-52 (Table 2).

Early-onset CRC has been associated with certain pathological features such as poor tumor differentiation and signet ring cell formation53-55, which may reflect underlying molecular characteristics of early-onset CRC. Studies showed that signet ring cell carcinoma (defined as carcinoma with more than 50% signet ring cell component56,57) constituted 6.4% (65/1,016)48 and 6.1% (26/428)49 of early-onset CRCs in patients aged <30 years compared to 2.3% (101/4,334)48 and 2.4% (42/1,751)49 in those aged 30-39 years; 1.0% (49/4,774)49 in those aged 40-49 years; and only 1.0-1.6% of later-onset CRCs32,49,57.

Studies have shown that the prevalence of MSI-high tumors was 10-30% in early-onset CRC37,39-41, compared to approximately 15% in overall CRC40. A study of mostly stage IV CRC cases (n=2,583) reported that, compared to later-onset CRC, early-onset CRC had fewer BRAF c.1799T>A (p.V600E) mutations, more MSI-high tumors, and fewer somatic mutations involved in the MAPK (mitogen-activated protein kinase) signaling pathway49. Another study using mostly advanced CRCs (n=18,218)58 showed that, compared to later-onset CRC, early-onset CRC patients aged <40 years exhibited lower prevalence of APC mutation (66% vs. 80%) and KRAS mutation (46% vs. 52%) in non-MSI-high tumors; as for MSI-high CRC, patients under age 40 had lower BRAF mutation prevalence (5.2% vs. 49%) and higher prevalence of APC (70% vs. 34%) and KRAS mutations (49% vs. 24%)58. Other analyses revealed differences in chromosomal aberrations between early-onset and later-onset CRCs in overall CRC52 or the non-MSI-high subset51.

With regard to epigenetic changes, early-onset CRC has been associated positively with tumor hypomethylation of long-interspersed nucleotide elements-1 (LINE-1) and inversely with high-level CpG island methylator phenotype (CIMP)59-61.

The distributions of consensus molecular subtypes (CMS) of CRC62 may also differ by age of onset. The CMS system was defined by an international consortium62 to be a classification based on comprehensive gene expression profiling. Each CMS subtype is associated with following features: CMS1, MSI-high and strong immune reaction; CMS2, activation of WNT and MYC signaling pathways; CMS3, specific metabolic signature; and CMS4, epithelial-mesenchymal transition62. In a study using two independent cohorts, early-onset CRC patients (n=82) and ≥70-year-old CRC patients (n=260) showed higher prevalence of CMS1 (22-23%) and lower prevalence of CMS2 (43%) and CMS4 (20-22%) compared to CRC patients aged 50-69 years (n=284) (11% CMS1, 50% CMS2, and 27% CMS4)49. The fraction of CMS3 was similar (12-13%) in early-onset and later-onset CRC49. The high CMS1 fraction in early-onset CRC patients may reflect an enrichment of Lynch syndrome MSI-high cases, supported by an even higher CMS1 fraction (44%; 11 of 25) in patients aged <40 years49.

Treatment & prognosis

Prognostic associations of early-onset CRC (compared to later-onset CRC) are mixed (Table 3); some suggested worse63-66 survival among early-onset CRC patients, while others reported similar67-72 or better32,55,73-78 prognosis. Particularly, more aggressive treatment in early-onset CRC has consistently been documented32,55,70,72,75,78-80. In a study using the Military Health System, early-onset CRC patients received on average 2-8 times more courses of postoperative systemic chemotherapy than later-onset CRC patients (aged 65-75 years) across all stages72. It remains unclear whether aggressive treatment for early-onset CRC can improve survival55,81 or not32,72,79. Prognostic features, clinical management schemes, and predictive biomarkers for specific treatment regimens in early-onset CRC remain to be determined. Notably, findings across studies are often incomparable due to the heterogeneity in study designs and populations.

Table 3.

Previous studies of prognosis and treatment of early-onset colorectal cancer.

Ref
No.
Authors
(year)
Country No. of cases
(diagnosed age)
Prognosis / treatment
77 O'Connell et al. (2004) United States 1,334 EOCRC (20-40) vs 46,457 LOCRC (60-80) The 5- year CRC-related survival of young CRC vs older CRC patients was 93% vs 95% (p, NS) for stage I, 89% vs 83% (p = 0.01) for stage II, and 59% vs 57% (p, NS) for stage III: 18% vs 6% (p < 0.001) for stage IV.
75 Quah et al. (2007) United States 68 EOCRC (≤40) vs 1,259 older CRC (>40) in Stage I-III The 5-year CRC-related survival of young vs older CRC patients was 86% vs 87% (p, NS). The 5-year OS was higher in the EOCRC (84% vs 73%, p = 0.001).
Young patients were also more likely to receive adjuvant chemotherapy, especially in the stage II: 39% vs 14%, (p = 0.003).
71 McMillan et al. (2009) United Kingdom 99 EOCRC (<45) vs 1,978 older CRC (≥45) Compared with young CRC patients aged <45, older CRC patients aged ≥45 had no significant difference in their hazard ratios for cancer-specific survival.
64 Sultan et al. (2010) Jordan 159 EOCRC (4-20) in 550,622 CRC SEER: The 5-year relative survival of young CRC vs older CRC were 40% vs 60% (p < 0.001). The 10-year relative survival, 31% vs 54%.
67 Blanke et al. (2011) Multiple countries* 793 EOCRC vs 5,491 LOCRC in stage IV Results from phase III trials. Response rate and OS were similar in CRC patients aged <50 and ≥50 in stage IV; PFS was minimally worse in the younger cohort (6.0 v 7.5 months; p = 0.02).
78 Hubbard et al. (2012) United States 5,817 EOCRC vs 27,757 LOCRC Younger patients aged <40 or <50 years with stage II and III colon cancer had longer OS and DFS than older patients. Younger patients (aged <40 or <50) with stage II and III colon cancer derive similar benefit from adjuvant therapy as older patients.
73 Schellerer et al. (2012) Germany 244 EOCRC vs 1,718 LOCRC The 5- year CRC-related survival of EOCRC vs LOCRC patients was 89% vs 80% (p = 0.01) for M0 rectal cancer, 21% vs 12% (p = 0.06) for M0 rectal cancer, 84% vs 89% (p, NS) for M1 colon cancer, 13% vs 15% (p, NS) for M1 colon cancer.
70 Yang et al. (2012) China 530 EOCRC (≤44) vs. 2,626 CRC (>44) Compared to older patients, young patients received chemotherapy and died of cancer related causes. OS, DFS and cancer-specific survival of younger patients were comparable to older patients.
63 Lieu et al. (2014) United States 3,051 EOCRC in 20,023 stage IV CRC Relative to patients of middle age CRC, young CRC patients had 19% (95% CI, 7-33%) increased risk of death and 22% (95% CI, 10-35%) increased risk of progression. The age effect did not differ by sites of metastatic disease, year of enrollment, types of therapy received, or biomarker mutational status.
32 Kneuertz et al. (2015) United States 13,102 EOCRC (18-49) vs 37,007 LOCRC (65-75) The 5-year CRC-related survival of young vs older CRC patients was 91% vs 90% for stage II overall, 95% vs 95% for stage II low risk, and 88% vs 86% for stage II high risk.
Almost 6% of the patients with young-onset stage I received adjuvant chemotherapy. Adjuvant chemotherapy was administered for 51% of young patients and 9% of older patients.
81 Orsini et al. (2015) Netherlands 1,102 EOCRC (≤40) vs 35,954 CRC (41-70) Young age was a prognostic factor for better survival in stage I-III patients. Adjuvant chemotherapy was more often given to young CRC patients (24% vs 14%, p <0.001). Adjuvant chemotherapy in young stage III and pN1 patients was associated with improved survival.
74 Wang et al. (2015) Sweden and United States 43,821 EOCRC vs 466,113 LOCRC SEER: The 5-year relative survival of young CRC vs older CRC were 67% vs 61% (p <0.001) in SEER database; 75% vs 63% (p = 0.25) in Linköping Cancer database.
80 Abdelsattar et al. (2016) United States 37,847 EOCRC vs 220,177 LOCRC (50-75) SEER: CRC patients with distant metastasis were more likely to receive surgical therapy for their primary tumors in younger age group (adjusted probability: 72% vs 63%; p < 0.001), and radiation therapy was more likely in younger CRC patients (adjusted probability: 53% vs 48%; p <0.001).
76 Boyce et al. (2016) Australia 2,001 EOCRC vs 30,177 LOCRC 5-year cancer-specific survival was greater for patients with EOCRC than for LOCRC (69% vs 66%) (p <0.001).
29 Holowatyj et al. (2016) United States 28,145 EOCRC SEER: The 5-year OS was 55% among non-Hispanic blacks, 68% among non-Hispanic whites, and 63% among Hispanic individuals (p <0.001).
65 Kim et al. (2016) Korea 693 EOCRC (≤45) vs 1,823 LOCRC (56-65) The 5-year cancer-specific survival rate was 81% in the young group and 88% in the older group (p <0.001). No significant difference in OS for each stage.
66 Chou et al. (2017) Taiwan 3,395 EOCRC (≤40) in 61,789 CRC Young CRC (≤40) had poorer OS and cancer-specific survival compared to older CRC (41–50 and 51–60) groups, although this trend was reversed in the 71–80 and >80 age groups.
72 Manjelievskaia et al. (2017) United States 671 EOCRC, 1,599 LOCRC (50-64), and 873 LOCRC (≥65) EOCRC and middle-aged CRC patients were more likely to receive multiagent chemotherapy than older patients (EOCRC: OR, 2.5; 95% CI, 1.4-4.3 and middle-aged CRC: OR, 2.7; 95% CI, 1.7-4.2). Among patients who received surgery and postoperative systemic chemotherapy, no significant differences were observed in survival among age groups.
68 Pokharkar et al. (2017) India 351 EOCRC (≤45) vs 427 CRC (>45) No statistically significant difference was found in OS between young and old CRC patients.
69 Rho et al. (2017) Multiple countries** 224 EOCRC (≤44) vs 274 CRC (>44) The mortality analysis showed no significant differences between the two groups. The longitudinal treatment patterns for both groups were similar but more biological therapy was used for young CRC.
79 Kolarich et al. (2018) United States 9,126 EOCRC vs 33,980 LOCRC (50-75) The receiving rates of radiation therapy were 42 % in stage I EOCRC patients and 32% in LOCRC patients, respectively (p < 0.001); the receiving rates of chemoradiation therapy were 94% in stage II and III EOCRC patients and 88% in stage II and III LOCRC patients.
55 Rodriguez et al. (2018) Canada 107 EOCRC (≤40) vs. 5,364 LOCRC (>60) Compared with patients aged > 60 years, adjuvant chemotherapy was delivered more often to younger CRC patients aged ≤40 years for stage II (50% vs 13%; p <0.001) and stage III (> 92% vs 57%; p <0.001), respectively. OS and cancer-specific survival were superior for the patients aged ≤40 years compared with the patients aged >60 years.

Studies with 100 or fewer cases or cases overlapping with another study (eg, SEER database) may not be listed, except for some studies.

*

France, Germany, United Kingdom, and United States.

**

Bulgaria, Canada, Czech Republic, Georgia, Ireland, and Italy.

Abbreviations: CI, confidence interval; CRC, colorectal cancer; DFS, disease-free survival; EOCRC, early-onset colorectal cancer; LOCRC, later-onset colorectal cancer; NS, not significant; OR, odds ratio; OS, overall survival; PFS, progression-free survival; SEER, Surveillance, Epidemiology, and End Results.

Why is early-onset CRC increasing?

Epidemiological factors

It should be noted that the adulthood exposures to risk factors discussed in the preceding part cannot fully explain the recent rise of early-onset CRC. If those had been risk factors for early-onset CRC, the rise of early-onset CRC would have started much earlier around the 1950s-1960s (along with the rise of later-onset CRC8), when many western countries started experiencing substantial lifestyle changes and increased exposure to many established CRC risk factors. Hypothetically, the “westernizing” lifestyle changes (starting around the 1950s) might have affected individuals in the early stages of life in the 1950s to 1980s. Those individuals were young adults around the 1980s to 2010s, generally coinciding with the observed rise of early-onset CRC incidence. The hypothesis that early-life exposures are risk factors for early-onset CRC16,82 can explain the delayed rise in early-onset CRC incidence since the 1980s. In fact, the observed rise in early-onset CRC did not happen in the 1950s-1960s when the rise in later-onset CRC incidence started8. If this hypothesis is true, the reported associations of the adult exposures with early-onset CRC might possibly be due to unmeasured confounding by early-life exposures. Most previous epidemiological studies of early-onset CRC have utilized data on adulthood exposures but generally lack early-life information, which precludes evaluation of confounding by early-life exposures.

There has already been evidence for early-life exposures as risk factors for CRC83-87 including early-onset CRC24. Analyses using the NIH AARP Diet and Health Study83 and the Boyd Orr Cohort84 suggested that childhood and adolescent diet was associated with the risk of CRC in general. Analyses using the Nurses’ Health Study II (NHS2)24 and a prospective cohort of 230,000 Norwegian adolescents85 showed that adolescent obesity was associated with increased incidence of early-onset CRC and colon cancer, respectively. Energy restriction during childhood and adolescent (due to the Dutch Hunger Winter) was associated with lower incidence of CRC87. In addition, studies of childhood radiation exposure caused by atomic bombings in Japan88, the Chernobyl nuclear accident85, and childhood radiotherapies89 provide evidence for early-life insults as risk factors of later cancers. However, studies on early-life exposures and adult cancers such as early-onset CRC remain scarce.

The importance of early-life information in early-onset CRC research should be recognized. The life course of each individual starts from conception and encompasses the prenatal period, infancy, childhood, adolescence, and adulthood. Early life is characterized by major physiological and metabolic changes90,91. Considering the acceleration of cell division and turnover, early life may represent periods of higher susceptibility to adverse effects from risk factors linked with hyperinsulinemia, increased growth factor levels, DNA damage, inefficient DNA repair, and altered microbiome. Conceivably, changes in physiology during growth in early life may cause time-varying cellular vulnerability to insults (risk factors).

Considering the possibly decades-long latency in the transition from normal cells to malignant neoplasm92,93 and the salient birth cohort effect observed with early-onset CRC incidence, it would be prudent to evaluate early-life exposures in relation to future risk of early-onset CRC. Around the 1950s, a global shift started in diet toward higher consumption of processed meat, fast foods, edible oils, refined grains, high-fructose corn syrup, and sugar94,95. The global prevalence of childhood and adolescent obesity has increased more than five-fold in recent decades96, along with reductions in physical activity94. Exposure to substances such as antibiotics was increasingly more prevalent in the past several decades97,98. Prenatal and perinatal practices have also changed with increased use of reproductive technologies, cesarean sections, and bottle feeding, which might have unforeseen long-term effects on offspring99-101. In adolescents, the incidence of inflammatory bowel disease (IBD), an established CRC risk factor, has increased dramatically (average annual percent changes in incidence: Crohn’s disease, 4.3% during 1988-2011; ulcerative colitis, 2.7% during 1988-1999 and 11% during 2000-2011)102,103. The incidence of type 2 diabetes mellitus has sharply increased by 7.1% annually during 2002-2012 in youths aged 10-19 years104.

There exists a continuum of cancer predisposition from high-penetrance genetic conditions to moderately-penetrant variants plus/minus interactive influences of environment and low-penetrant variants to low-penetrant polygenic conditions with larger contributions of gene-by-environment (GxE) interactions. Near 100% penetrance risk variants for hereditary CRC, which certainly cause early-onset CRC, unlikely fully explain its recent rise. In contrast, low to moderately-penetrant variants with GxE interactions (especially in early-life) might contribute to the increase of early-onset CRC.

In addition, considering the apparent health disparities related to early-onset CRC26-28, future studies should investigate how race/ethnicity, other sociodemographic factors, and their interplay relate to early-life exposures and the etiology of early-onset CRC.

Characterizing the generational shifts in early-life exposures may provide insights into the etiologies of early-onset CRC as well as childhood and other early-onset cancers. Figure 1 depicts several points for better understanding of the etiology of early-onset CRC.

Figure 1.

Figure 1.

Illustration of potential effects of various life-course exposures on tumor development from early life (the prenatal to adolescent periods) to adulthood. The exposome indicates the totality of exposures and interactions thereof. The gene-by-environment (GxE) interactions during life-course may play an important role in the etiology of early-onset colorectal cancer. Early-life biospecimens such as stool, blood, saliva, urine, cord blood, placenta, etc., which can be collected from either mother or fetus/neonate or both, may be able to provide early-life information when analyzed in the future.

Abbreviations: GxE, gene-by-environment; wt, weight.

Tumor molecular features

Accumulating evidence indicates that exposure to diet, chemicals, and environmental factors may cause cellular epigenetic and genetic alterations105, leading to certain tumor molecular subtypes106. Compared to unexposed children and adolescents, those who had experienced famine during the “Dutch Hunger Winter” experienced lower incidence of CIMP-positive CRC but not CIMP-negative CRC86. Epigenetic and genetic alterations caused by certain early-life exposures might potentially contribute to the increase in early-onset CRC incidence.

LINE-1 hypomethylation, a surrogate marker of genome-wide DNA hypomethylation status, is occasionally observed in CRC59,60,107. A variety of exposures including ionizing radiation108, high BMI109, physical inactivity109, cigarette smoking110, pesticides110, benzene110, etc. have been associated with lower LINE-1 methylation levels in blood cells. Considering the association between younger age at CRC diagnosis and tumor LINE-1 hypomethylation59,61, these exposures (together with gene-by-environment interactions), especially in early life, may play an etiological role in early-onset CRC.

A recent experimental analysis using paired normal and early-onset CRC organoid models has demonstrated its potential to reveal novel signaling pathways important in pathogenesis of early-onset CRC111.

Microbiome & immunity

Ample evidence indicates that the gut microbiota may contribute to colorectal tumor evolution112-114, and several species such as Fusobacterium nucleatum115,116, Escherichia coli carrying the polyketide synthase (pks) island117, and enterotoxigenic Bacteroides fragilis118 as well as overall dysbiosis have been implicated112-114. The composition of microorganisms changes according to the anatomical location in the colorectum119, which may relate to the epidemiological, microbial, and tumor molecular differences in CRC according to detailed colorectal subsites8,46,47,120. Although F. nucleatum may contribute to CRC development via its effects on cell proliferation and anti-tumor immunity121, the link between F. nucleatum and early-onset CRC remains underexplored. In addition, Bifidobacterium in CRC tissue has been associated with signet ring cell formation122, a feature that has been associated with early-onset CRC48,49.

Prenatal, perinatal, and neonatal exposures such as maternal diabetes123, alcohol20, smoking123, cesarean delivery124, bottle feeding124, and antibiotics use125-127 may influence the developing microbiota and immune system. Accumulating evidence indicates that diverse immune cell types develop and mature at different gestational stages, and that these processes are essential in establishing immune tolerance and response according to developmental needs128. With regard to increased antibiotic use which may result in systemic and intestinal dysbiosis throughout the life course, studies have shown an association of long-term antibiotic use with CRC and colorectal adenoma129-131. Any ingested substances including foods, antibiotics, and chemicals (such as colorants and preservatives in modern food products132) may possibly alter intestinal microbes and their metabolisms and/or could be metabolized by the microbes, which can then affect fetal physiology via umbilical circulation and amniotic fluids133. The long-term impact of these ingestants and altered microbial features in pregnancy and early life needs to be studied.

It is challenging to decipher the complex interactions among exposures (diet, antibiotics, chemicals, smoking, alcohol, etc.), microbiome, and host, all of which may play a significant role in CRC etiology. To examine these interactions in early life, we need prospective (and ideally longitudinal) collections of early-life information and biospecimens such as stool from pregnant mothers and children.

Primary & secondary prevention

The impact of early-onset CRC prevention in young individuals, if successful, will be substantial, considering the additional decades of life expectancy that would result. As the avoidance of risk factors is always a major primary prevention measure, further research to establish the risk factors of early-onset CRC is needed. In addition, it is recommended that early-onset CRC patients with high-level MSI or MMR protein loss undergo germline MMR gene sequencing to identify Lynch syndrome cases for prevention of future CRC in family members42.

Although screening with stool-based tests and colonoscopy may be useful for secondary prevention of early-onset CRC, methods of screening as well as the recommended age to start screening in average-risk or higher-risk persons have yet to be widely agreed upon134,135. A recent systematic review on the prevalence of adenoma in colonoscopy populations, based on 19 studies comprising 19,295 individuals, reported the summary prevalence of early-onset adenoma of 11% (95% confidence interval, 8.5%−14%)136. The U.S. Multi-Society Task Force recommends that persons with a family history of CRC in a first-degree relative diagnosed before 60 years of age undergo colonoscopy every 5 years beginning at age 40 or 10 years before the age the relative was diagnosed, whichever is earlier137. In 2018, the American Cancer Society (ACS) recommended that screening start at age 45 years for average-risk persons138. In a nationally representative sample of individuals aged 45-49 years, past-year CRC screening rates increased from 4.8% to 12% within 2018, coinciding with the ACS guideline release139. The 2018 ACS recommendation to begin screening at age 45 years is based on disease burden, results from microsimulation modeling, and the reasonable expectation that screening will perform similarly in adults aged 45 to 49 years as older population138. Similar to prior modeling work to support the U.S. Preventive Service Task Force guidelines140, the microsimulation modeling studies used to support the ACS guideline captured 1) observed/projected changes in cancer incidence; 2) benefits/harms of each screening modality; and 3) balance between benefits and the burden of screening. Specifically, the modeling suggests that screening every 10 years from ages 45 to 75 years achieved a ratio of incremental burden (number of colonoscopies) to benefit (life-years gained) of 32. This strategy was selected because it was on the efficient frontier and had the highest number of life-years gained among the strategies with ratios of less than 45141.

Although screening should ideally be tailored for each person according to one’s own risk, individualized risk assessment has not yet been adopted to CRC screening recommendations. It is likely that the assessment of genetic germline polymorphisms, early-life exposures, and their GxE interactions will improve the efficiency of screening and early-detection of early-onset CRC. However, evidence is scarce and additional studies are needed.

Transdisciplinary epidemiology

Integration of molecular biologic methods into epidemiology has become increasingly common under the umbrella term of molecular epidemiology since the 1980s106. More recently, the field of molecular pathological epidemiology (MPE) has substantially grown along with the increasing availability of tumor tissue resources and molecular assays in population-based studies106,142. The strengths of MPE research have been most apparent when an association between an exposure and a disease entity has not been established with certainty. Using the MPE approach, a putative risk factor can be linked to molecular pathology, which can provide insight into etiological mechanisms106,142. Moreover, when such an etiological link exists, we can expect to observe a stronger association with the specific disease subtype than with the overall disease entity that contains pathogenetically heterogeneous subtypes106,142. Therefore, MPE research can contribute to the establishment of causality106,142. Recently, the concept of MPE has been increasingly recognized in the literature143-152. While somewhat overlapping with the concepts of “molecular epidemiology” and “systems epidemiology”, MPE places particular emphasis on tumor tissue analyses for better pathobiological understanding.

MPE can play an important role in early-onset CRC research due to its ability to unmask previously unknown risk factors and establish causality. If a putative risk factor can be linked to specific pathogenic signatures, the MPE approach can support it as a new risk factor for early-onset CRC. Through the discovery of hidden risk factors, MPE research can inform the development of primary prevention strategies for early-onset CRC.

Challenges & opportunities

Epidemiological studies of adulthood exposures and early-onset CRC have thus far not successfully identified the root causes of its recent rise. As already discussed, if adulthood exposures were indeed risk factors for early-onset CRC, we would have observed the rise of early-onset CRC incidence even in the 1950s similar to the rise in later-onset CRC incidence. One of the major challenges in existing studies of early-onset CRC is the severe paucity of reliable early-life data. Notably, much early-life information needs to be obtained from early-life biospecimens or parents (especially mothers).

To expedite research on early-onset cancers and examine early-life risk factors, existing large-scale prospective cohort studies can be utilized. The Growing Up Today Study (GUTS)153 has been following 16,882 children (9 to 14 years old at the time of enrollment in 1996) of female participants in the Nurses’ Health Study II (NHS2). The GUTS2 started to follow an additional cohort of 10,923 children (10 to 17 years old at the time of enrollment in 2004) of the NHS2 participants. The NHS224, the parent cohort of these child cohorts, has been following 116,430 participating women who were 25 to 42 years old at the time of the study initiation in 1989. The NHS2 has prospectively collected data on exposures and various outcomes, including reproductive health and chronic diseases such as cancer. Cohorts of parent-offspring pairs can provide information in the prenatal, perinatal, neonatal, and childhood periods, which may contribute novel evidence to the study of the role of various early-life exposures in diseases during the life course. The NHS3 (https://www.nhs3.org/), which includes 45,000 U.S. nurses and nursing students aged 18 and above, recently started another recruitment phase to reach a target population of 100,000.

The Colon Cancer Family Registry (CCFR) Cohort154, which includes 42,489 participants from 15,049 families, was established in 1997 as an international consortium of six centers in North America and Australia. The CCFR collected information regarding family history of cancer and CRC risk factors, as well as biospecimens (blood and paraffin-embedded tumor tissue). During phase II recruitment (2002-2007), the population-based recruitment efforts targeted early-onset CRC cases. They have identified over 3,000 early-onset CRC cases154. In the future, this study will contribute to the clarification of the genetic and environmental etiologies of early-onset CRC.

The Child Health and Development Studies (CHDS)155, which includes over 15,000 women and their 19,000 children, was initiated in 1959 to investigate the associations of biological, genetic, and early-life factors with health outcomes in adults. The biospecimens and data on early-life to adult exposures collected over the past 60 years will provide us with a unique resource to evaluate the relationship between early-life exposures and long-term health outcomes.

The National Children’s Study (https://www.nichd.nih.gov/research/supported/NCS) was an ambitious project with goals to recruit and follow 100,000 children in the U.S. from prenatal to adulthood periods, and to study environmental influences on child health and development. Although the study was closed in 2014, a reactivation of the study or following-up of the former participants has been suggested156, which may provide invaluable opportunities to study the etiologies of childhood and early-onset cancers.

Growing Up in New Zealand157 is a longitudinal study that includes more than 6,000 children and their families. This study was designed to recruit and follow a cohort of children from the prenatal period onward. Findings from this study may provide yet more evidence on early-life exposures and early-onset cancers.

There are several other studies that have the potential to provide new insight into early-onset CRC156; those include the UK Millennium Cohort Study158 and the ORIGINS Project (https://originsproject.telethonkids.org.au/) in Australia. These studies will provide opportunities to clarify the effect of early-life exposures on long-term health outcomes including cancer.

Importantly, these valuable datasets and any other relevant studies may be harmonized, combined, and/or integrated to increase the validity of findings and illuminate further information on the etiologies of a multitude of health conditions throughout the life course, including (but not limited to) early-onset CRC.

Conclusion & future direction

Although the causes of the rise of early-onset CRC remain uncertain, we can deploy and implement several measures relatively fast. Those include (1) education of physicians, other healthcare workers, and general individuals; (2) re-evaluation of screening guidelines for early-onset CRC; (3) etiological research for identifying genetic and epidemiologic risk factors; and (4) better coordination of clinical care and communication for early-onset CRC patients82. We advocate the roadmap of the general strategies in Table 4. Education of healthcare providers, patients, and general individuals regarding the rise of early-onset CRC may lead to prevention and early detection. It would be ideal that researchers learn the importance of longitudinal life-course cohort studies combined with prospective biospecimen collections.

Table 4.

Roadmap of strategies to reduce the public health burden of early-onset CRC.

Theme Strategy Note
1: Education 1.1: Educate physicians, other healthcare workers, and general individuals to raise awareness of early-onset CRC. This can be implemented.
2: Screening 2.1: Expand genetic testing for individuals with family history of CRC.
2.2: Conduct research to create and refine prediction models for early-onset CRC.
2.3: Evaluate available evidence and screening guidelines for early-onset CRC.
2.4: Develop personalized screening strategies.
Further research, including comparative effectiveness research, is needed.
3: Etiological research 3.1: Utilize existing resources to study etiologies.
3.2: Design additional studies, including life-course cohort studies, combined with biospecimen collections and omics analyses.
3.3: Study genetic and epidemiologic risk factors, including early-life exposures, (and GxE interactions) for early-onset CRC.
3.4: Study etiological mechanisms using experimental model systems.
Determination of risk factors and their effect sizes can help us improve primary, secondary, and tertiary prevention strategies.
4: Clinical care and research 4.1: Set up specialized centers, units, and/or clinics focused on early-onset CRC, to deliver improved care.
4.2: Conduct clinical trials to determine personalized treatment strategies.
4.3: Communicate with early-onset CRC patients regarding treatment options and considerations as well as implications for potential familial risks.
Randomized clinical trials integrated with assessments of tumor characteristics (including tissue microbiota and immunity) are needed. Genetic, fertility, and social counseling may be indicated for patients.

Abbreviation: CRC, colorectal cancer; GxE, gene-by-environment.

Thus far, attempts to identify adulthood exposures as risk factors for early-onset CRC have seen only limited success. Clues may emerge when we carefully consider (1) the likely long (years to decades) growth / incubation time of colorectal neoplasms; (2) the environmental, dietary, and lifestyle changes that have occurred over the past century; (3) the observed birth cohort effects; (4) the drastic biophysiological changes that occur in each individual from conception to adulthood; and (5) the differential clinical, pathological, and molecular features of early-onset CRC compared to later-onset CRC. It is conceivable that currently unknown etiological factors, possibly those that begin in early life (from the prenatal to adolescent periods), may be driving the rise of early-onset CRC incidence. Challenges include the paucity of large-scale longitudinal studies that can provide accurate information on long-term exposures from early life to adulthood. We must develop life-course epidemiological studies, which should be combined with prospective biospecimen collections (stool, blood, saliva, urine, placenta, amniotic fluid, cord blood, etc.), state-of-the-art omics (including genomic, metagenomic, and metabolomic research), and analyses of molecular pathology, immunity, and the tumor microenvironment159. Importantly, such prospective cohorts, once established, can be utilized to elucidate the etiologies of not only early-onset CRC but also many other diseases including childhood cancers and other early-onset malignancies. We can consider a following scenario: if we have resources, should we invest to establish (A) 10 parent-offspring paired prospective cohorts vs. (B) 100 case-control studies? In the former, we can study not only early-life exposures but also dozens of diseases of children and adults. In the latter, each study can assess only one disease of interest, and cannot fully evaluate early-life exposures from adult subjects, such as early-onset CRC patients. It is quite evident which can benefit us more in the long run. Hence, prudent investment of our finite resources, funds, and efforts into building longitudinal prospective cohort infrastructure including biobanks should be our key priority.

In the meantime, astute use of existing datasets and resources including large prospective cohort studies to examine early-life information offers a relatively cost- and time-efficient means to expedite etiological research on early-onset CRC and other early-onset cancers.

Acknowledgements:

The authors thank Mr. Tyler Twombly for proof reading of the manuscript. This work was supported by U.S. National Institutes of Health (NIH) grants (R35 CA197735 to S.O.; R21 CA230873 to S.O. and K.W.; R01 CA248857 to S.O.; R01 CA205406 to K.N.; R03 CA197879 to K.W.; R37 CA246175 to Y.C.); by Cancer Research UK Grand Challenge Award (UK C10674/A27140 to M.G., K.N., and S.O.); by an Investigator Initiated Grant from the American Institute for Cancer Research (AICR) to K.W.; and by U.S. Department of Defense (CA160344 to K.N.). K.N. and M.G. had received support from the Project P Fund. M.G. was supported by the Stand Up to Cancer Colorectal Cancer Dream Team Translational Research Grant (SU2C-AACR-DT22-17) administered by the American Association for Cancer Research, a scientific partner of SU2C. T.U. was supported by a grant from Overseas Research Fellowship (201960541) from Japan Society for the Promotion of Science, the Uehara Memorial Foundation, and Yasuda Medical Foundation. K.F. were supported by fellowship grants from the Uehara Memorial Foundation and the Grant of The Clinical Research Promotion Foundation (2018). The content is solely the responsibility of the authors and does not necessarily represent the official views of NIH. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Abbreviations:

ACS

American Cancer Society

AJCC

American Joint Committee on Cancer

BMI

body mass index

CCFR

Colon Cancer Family Registry

CHDS

Child Health and Development Studies

CIMP

CpG island methylator phenotype

CMS

consensus molecular subtype

CRC

colorectal cancer

FAP

familial adenomatous polyposis

GxE

gene-by-environment

GUTS

Growing Up Today Study

IBD

inflammatory bowel disease

LINE-1

long interspersed nucleotide element-1

MMR

mismatch repair

MPE

molecular pathological epidemiology

MSI

microsatellite instability

NHS2

Nurses’ Health Study II

NHS3

Nurses’ Health Study 3

pks

polyketide synthase

Footnotes

Competing interests: K.N. has received institutional research funding from Pharmavite, Revolution Medicines, Evergrande Group, Genentech, Gilead Sciences, Trovagene, and Tarrex Biopharma; and served on advisory boards for Bayer, Seattle Genetics, and Array Biopharma. M.G. has received research funding from Bristol-Myers Squibb and Merck. The other authors declare that they have no conflicts of interest.

Use of standardized official symbols: We use HUGO (Human Genome Organisation)-approved official symbols (or root symbols) for genes and gene products, including APC, ATM, BRAF, BRCA1, BRCA2, CDKN2A, CHECK2, CTNNB1, KRAS, MAPK, MLH1, MSH2, MSH6, MUTYH, PALB2, PMS2, SMAD4, and TP53; all of which are described at www.genenames.org. Gene symbols are italicized whereas symbols for gene products are not italicized.

References:

  • 1.Siegel RL et al. Colorectal cancer statistics, 2020. CA Cancer J Clin Epub ahead of print (2020). [Google Scholar]
  • 2.Siegel RL et al. Global patterns and trends in colorectal cancer incidence in young adults. Gut 68, 2179–2185 (2019). [DOI] [PubMed] [Google Scholar]
  • 3.Vuik FE et al. Increasing incidence of colorectal cancer in young adults in Europe over the last 25 years. Gut 68, 1820–1826 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bray F et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68, 394–424 (2018). [DOI] [PubMed] [Google Scholar]
  • 5.Arnold M et al. Global patterns and trends in colorectal cancer incidence and mortality. Gut 66, 683–691 (2017). [DOI] [PubMed] [Google Scholar]
  • 6.Global Burden of Disease Cancer, C. et al. Global, Regional, and National Cancer Incidence, Mortality, Years of Life Lost, Years Lived With Disability, and Disability-Adjusted Life-years for 32 Cancer Groups, 1990 to 2015: A Systematic Analysis for the Global Burden of Disease Study. JAMA Oncol 3, 524–548 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Dekker E, Tanis PJ, Vleugels JLA, Kasi PM & Wallace MB Colorectal cancer. Lancet 394, 1467–1480 (2019). [DOI] [PubMed] [Google Scholar]
  • 8.Keum N & Giovannucci E Global burden of colorectal cancer: emerging trends, risk factors and prevention strategies. Nat Rev Gastroenterol Hepatol 16, 713–732 (2019). [DOI] [PubMed] [Google Scholar]
  • 9.Lui RN et al. Global Increasing Incidence of Young-Onset Colorectal Cancer Across 5 Continents: A Joinpoint Regression Analysis of 1,922,167 Cases. Cancer Epidemiol Biomarkers Prev 28, 1275–1282 (2019). [DOI] [PubMed] [Google Scholar]
  • 10.Saad El Din K et al. Trends in the epidemiology of young-onset colorectal cancer: a worldwide systematic review. BMC Cancer 20, 288 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Meester RGS, Mannalithara A, Lansdorp-Vogelaar I & Ladabaum U Trends in Incidence and Stage at Diagnosis of Colorectal Cancer in Adults Aged 40 Through 49 Years, 1975-2015. JAMA 321, 1933–1934 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Siegel RL et al. Colorectal Cancer Incidence Patterns in the United States, 1974-2013. J Natl Cancer Inst 109 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Chung RY et al. A population-based age-period-cohort study of colorectal cancer incidence comparing Asia against the West. Cancer Epidemiol 59, 29–36 (2019). [DOI] [PubMed] [Google Scholar]
  • 14.Brenner DR et al. Increasing colorectal cancer incidence trends among younger adults in Canada. Prev Med 105, 345–349 (2017). [DOI] [PubMed] [Google Scholar]
  • 15.Feletto E et al. Trends in Colon and Rectal Cancer Incidence in Australia from 1982 to 2014: Analysis of Data on Over 375,000 Cases. Cancer Epidemiol Biomarkers Prev 28, 83–90 (2019). [DOI] [PubMed] [Google Scholar]
  • 16.Stoffel EM & Murphy CC Epidemiology and Mechanisms of the Increasing Incidence of Colon and Rectal Cancers in Young Adults. Gastroenterology 158, 341–353 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Sung JJY et al. Increasing Trend in Young-Onset Colorectal Cancer in Asia: More Cancers in Men and More Rectal Cancers. Am J Gastroenterol 114, 322–329 (2019). [DOI] [PubMed] [Google Scholar]
  • 18.Sung H, Siegel RL, Rosenberg PS & Jemal A Emerging cancer trends among young adults in the USA: analysis of a population-based cancer registry. Lancet Public Health 4, e137–e147 (2019). [DOI] [PubMed] [Google Scholar]
  • 19.Gupta S et al. International trends in the incidence of cancer among adolescents and young adults. J Natl Cancer Inst Epub ahead of print (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Rosato V et al. Risk factors for young-onset colorectal cancer. Cancer Causes Control 24, 335–341 (2013). [DOI] [PubMed] [Google Scholar]
  • 21.Gausman V et al. Risk Factors Associated With Early-Onset Colorectal Cancer. Clin Gastroenterol Hepatol (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Low EE et al. Risk Factors for Early-Onset Colorectal Cancer. Gastroenterology Epub ahead of print (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Nguyen LH et al. Sedentary Behaviors, TV Viewing Time, and Risk of Young-Onset Colorectal Cancer. JNCI Cancer Spectr 2, pky073 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Liu PH et al. Association of Obesity With Risk of Early-Onset Colorectal Cancer Among Women. JAMA Oncol 5, 37–44 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Liang PS, Mayer JD, Wakefield J & Ko CW Temporal Trends in Geographic and Sociodemographic Disparities in Colorectal Cancer Among Medicare Patients, 1973-2010. J Rural Health 33, 361–370 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Murphy CC, Wallace K, Sandler RS & Baron JA Racial Disparities in Incidence of Young-Onset Colorectal Cancer and Patient Survival. Gastroenterology 156, 958–965 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Mayer-Davis EJ et al. Incidence Trends of Type 1 and Type 2 Diabetes among Youths, 2002-2012. N Engl J Med 376, 1419–1429 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Ogden CL et al. Trends in Obesity Prevalence Among Children and Adolescents in the United States, 1988-1994 Through 2013-2014. JAMA 315, 2292–2299 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Holowatyj AN, Ruterbusch JJ, Rozek LS, Cote ML & Stoffel EM Racial/Ethnic Disparities in Survival Among Patients With Young-Onset Colorectal Cancer. J Clin Oncol 34, 2148–2156 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Archambault AN et al. Cumulative Burden of Colorectal Cancer-Associated Genetic Variants Is More Strongly Associated With Early-Onset vs Late-Onset Cancer. Gastroenterology 158, 1274–1286.e1212 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wang L et al. Risk Factor Profiles Differ for Cancers of Different Regions of the Colorectum. Gastroenterology Epub ahead of print (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kneuertz PJ et al. Overtreatment of young adults with colon cancer: more intense treatments with unmatched survival gains. JAMA Surg 150, 402–409 (2015). [DOI] [PubMed] [Google Scholar]
  • 33.Myers EA et al. Colorectal cancer in patients under 50 years of age: a retrospective analysis of two institutions' experience. World J Gastroenterol 19, 5651–5657 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Chen FW, Sundaram V, Chew TA & Ladabaum U Advanced-Stage Colorectal Cancer in Persons Younger Than 50 Years Not Associated With Longer Duration of Symptoms or Time to Diagnosis. Clin Gastroenterol Hepatol 15, 728–737 e723 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Deng SX et al. Factors influencing diagnosis of colorectal cancer: a hospital-based survey in China. J Dig Dis 13, 517–524 (2012). [DOI] [PubMed] [Google Scholar]
  • 36.Ben-Ishay O et al. Diagnosis of colon cancer differs in younger versus older patients despite similar complaints. Isr Med Assoc J 15, 284–287 (2013). [PubMed] [Google Scholar]
  • 37.Liang JT et al. Clinicopathological and molecular biological features of colorectal cancer in patients less than 40 years of age. Br J Surg 90, 205–214 (2003). [DOI] [PubMed] [Google Scholar]
  • 38.Siegel RL, Jakubowski CD, Fedewa SA, Davis A & Azad NS Colorectal Cancer in the Young: Epidemiology, Prevention, Management. Am Soc Clin Oncol Educ Book 40, 1–14 (2020). [DOI] [PubMed] [Google Scholar]
  • 39.Stoffel EM et al. Germline Genetic Features of Young Individuals With Colorectal Cancer. Gastroenterology 154, 897–905.e891 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Pearlman R et al. Prevalence and Spectrum of Germline Cancer Susceptibility Gene Mutations Among Patients With Early-Onset Colorectal Cancer. JAMA Oncol 3, 464–471 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Mork ME et al. High Prevalence of Hereditary Cancer Syndromes in Adolescents and Young Adults With Colorectal Cancer. J Clin Oncol 33, 3544–3549 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Valle L, Vilar E, Tavtigian SV & Stoffel EM Genetic predisposition to colorectal cancer: syndromes, genes, classification of genetic variants and implications for precision medicine. J Pathol 247, 574–588 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Koskenvuo L, Ryynänen H & Lepistö A Timing of prophylactic colectomy in familial adenomatous polyposis. Colorectal Dis Epub ahead of print (2020). [DOI] [PubMed] [Google Scholar]
  • 44.Ogino S et al. Integrative analysis of exogenous, endogenous, tumour and immune factors for precision medicine. Gut 67, 1168–1180 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Yamauchi M et al. Assessment of colorectal cancer molecular features along bowel subsites challenges the conception of distinct dichotomy of proximal versus distal colorectum. Gut 61, 847–854 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Yamauchi M et al. Colorectal cancer: a tale of two sides or a continuum? Gut 61, 794–797 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mima K et al. Fusobacterium nucleatum in Colorectal Carcinoma Tissue According to Tumor Location. Clin Transl Gastroenterol 7, e200 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Holowatyj AN et al. Clinicopathologic and Racial/Ethnic Differences of Colorectal Cancer Among Adolescents and Young Adults. Clin Transl Gastroenterol 10, e00059 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Willauer AN et al. Clinical and molecular characterization of early-onset colorectal cancer. Cancer 125, 2002–2010 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Perea J et al. Age at onset should be a major criterion for subclassification of colorectal cancer. J Mol Diagn 16, 116–126 (2014). [DOI] [PubMed] [Google Scholar]
  • 51.Berg M et al. Distinct high resolution genome profiles of early onset and late onset colorectal cancer integrated with gene expression data identify candidate susceptibility loci. Mol Cancer 9, 100 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Arriba M et al. DNA copy number profiling reveals different patterns of chromosomal instability within colorectal cancer according to the age of onset. Mol Carcinog 55, 705–716 (2016). [DOI] [PubMed] [Google Scholar]
  • 53.Yeo H et al. Early-onset Colorectal Cancer is Distinct From Traditional Colorectal Cancer. Clin Colorectal Cancer 16, 293–299.e296 (2017). [DOI] [PubMed] [Google Scholar]
  • 54.Soliman BG, Karagkounis G, Church JM, Plesec T & Kalady MF Mucinous Histology Signifies Poor Oncologic Outcome in Young Patients With Colorectal Cancer. Dis Colon Rectum 61, 547–553 (2018). [DOI] [PubMed] [Google Scholar]
  • 55.Rodriguez L et al. Disease Characteristics, Clinical Management, and Outcomes of Young Patients With Colon Cancer: A Population-based Study. Clin Colorectal Cancer 17, e651–e661 (2018). [DOI] [PubMed] [Google Scholar]
  • 56.Nagtegaal I, Arends M & Salto-Tellez M in WHO Classification of Tumours, 5th edition: Digestive System Tumours (ed WHO Classification of Tumours Editorial Board) 180 (World Health Organization, Geneva, 2019). [Google Scholar]
  • 57.Inamura K et al. Prognostic significance and molecular features of signet-ring cell and mucinous components in colorectal carcinoma. Ann Surg Oncol 22, 1226–1235 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Lieu CH et al. Comprehensive Genomic Landscapes in Early and Later Onset Colorectal Cancer. Clin Cancer Res 25, 5852–5858 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Antelo M et al. A high degree of LINE-1 hypomethylation is a unique feature of early-onset colorectal cancer. PLoS One 7, e45357 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Feinberg AP The Key Role of Epigenetics in Human Disease Prevention and Mitigation. N Engl J Med 378, 1323–1334 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Baba Y et al. Epigenomic diversity of colorectal cancer indicated by LINE-1 methylation in a database of 869 tumors. Mol Cancer 9, 125 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Guinney J et al. The consensus molecular subtypes of colorectal cancer. Nat Med 21, 1350–1356 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lieu CH et al. Association of age with survival in patients with metastatic colorectal cancer: analysis from the ARCAD Clinical Trials Program. J Clin Oncol 32, 2975–2984 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Sultan I et al. Distinct features of colorectal cancer in children and adolescents: a population-based study of 159 cases. Cancer 116, 758–765 (2010). [DOI] [PubMed] [Google Scholar]
  • 65.Kim TJ, Kim ER, Hong SN, Chang DK & Kim YH Long-Term Outcome and Prognostic Factors of Sporadic Colorectal Cancer in Young Patients: A Large Institutional-Based Retrospective Study. Medicine (Baltimore) 95, e3641 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chou CL, Tseng CJ & Shiue YL The impact of young age on the prognosis for colorectal cancer: a population-based study in Taiwan. Jpn J Clin Oncol 47, 1010–1018 (2017). [DOI] [PubMed] [Google Scholar]
  • 67.Blanke CD et al. Impact of young age on treatment efficacy and safety in advanced colorectal cancer: a pooled analysis of patients from nine first-line phase III chemotherapy trials. J Clin Oncol 29, 2781–2786 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Pokharkar AB et al. Young Vs Old Colorectal Cancer in Indian Subcontinent: a Tertiary Care Center Experience. Indian J Surg Oncol 8, 491–498 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Rho YS et al. Comparing Clinical Characteristics and Outcomes of Young-onset and Late-onset Colorectal Cancer: An International Collaborative Study. Clin Colorectal Cancer 16, 334–342 (2017). [DOI] [PubMed] [Google Scholar]
  • 70.Yang Z et al. Characteristics and long-term survival of colorectal cancer patients aged 44 years and younger. Clin Transl Oncol 14, 896–904 (2012). [DOI] [PubMed] [Google Scholar]
  • 71.McMillan DC & McArdle CS The impact of young age on cancer-specific and non-cancer-related survival after surgery for colorectal cancer: 10-year follow-up. Br J Cancer 101, 557–560 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Manjelievskaia J et al. Chemotherapy Use and Survival Among Young and Middle-Aged Patients With Colon Cancer. JAMA Surg 152, 452–459 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Schellerer VS et al. Despite aggressive histopathology survival is not impaired in young patients with colorectal cancer : CRC in patients under 50 years of age. Int J Colorectal Dis 27, 71–79 (2012). [DOI] [PubMed] [Google Scholar]
  • 74.Wang MJ et al. The prognostic factors and multiple biomarkers in young patients with colorectal cancer. Sci Rep 5, 10645 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Quah HM et al. Young age influences treatment but not outcome of colon cancer. Ann Surg Oncol 14, 2759–2765 (2007). [DOI] [PubMed] [Google Scholar]
  • 76.Boyce S et al. Young-onset colorectal cancer in New South Wales: a population-based study. Med J Aust 205, 465–470 (2016). [DOI] [PubMed] [Google Scholar]
  • 77.O'Connell JB et al. Do young colon cancer patients have worse outcomes? World J Surg 28, 558–562 (2004). [DOI] [PubMed] [Google Scholar]
  • 78.Hubbard J et al. Benefits and adverse events in younger versus older patients receiving adjuvant chemotherapy for colon cancer: findings from the Adjuvant Colon Cancer Endpoints data set. J Clin Oncol 30, 2334–2339 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kolarich A et al. Rectal cancer patients younger than 50 years lack a survival benefit from NCCN guideline-directed treatment for stage II and III disease. Cancer 124, 3510–3519 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Abdelsattar ZM et al. Colorectal cancer outcomes and treatment patterns in patients too young for average-risk screening. Cancer 122, 929–934 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Orsini RG et al. Comparable survival for young rectal cancer patients, despite unfavourable morphology and more advanced-stage disease. Eur J Cancer 51, 1675–1682 (2015). [DOI] [PubMed] [Google Scholar]
  • 82.Hofseth LJ et al. Early-onset colorectal cancer: initial clues and current views. Nat Rev Gastroenterol Hepatol 17, 352–364 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Ruder EH et al. Adolescent and mid-life diet: risk of colorectal cancer in the NIH-AARP Diet and Health Study. Am J Clin Nutr 94, 1607–1619 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.van der Pols JC et al. Childhood dairy intake and adult cancer risk: 65-y follow-up of the Boyd Orr cohort. Am J Clin Nutr 86, 1722–1729 (2007). [DOI] [PubMed] [Google Scholar]
  • 85.Bjørge T, Engeland A, Tverdal A & Smith GD Body mass index in adolescence in relation to cause-specific mortality: a follow-up of 230,000 Norwegian adolescents. Am J Epidemiol 168, 30–37 (2008). [DOI] [PubMed] [Google Scholar]
  • 86.Hughes LA et al. Early life exposure to famine and colorectal cancer risk: a role for epigenetic mechanisms. PLoS One 4, e7951 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hughes LA et al. Childhood and adolescent energy restriction and subsequent colorectal cancer risk: results from the Netherlands Cohort Study. Int J Epidemiol 39, 1333–1344 (2010). [DOI] [PubMed] [Google Scholar]
  • 88.Boice JD Jr. in Cancer Epidemiology and Prevention (eds Schottenfeld D & Fraumeni JF Jr) 259–293 (Oxford University Press, New York, 2006). [Google Scholar]
  • 89.Sigurdson AJ et al. Primary thyroid cancer after a first tumour in childhood (the Childhood Cancer Survivor Study): a nested case-control study. Lancet 365, 2014–2023 (2005). [DOI] [PubMed] [Google Scholar]
  • 90.Caprio S et al. Central adiposity and its metabolic correlates in obese adolescent girls. Am J Physiol 269, E118–126 (1995). [DOI] [PubMed] [Google Scholar]
  • 91.Chiarelli F & Marcovecchio ML Insulin resistance and obesity in childhood. Eur J Endocrinol 159 Suppl 1, S67–74 (2008). [DOI] [PubMed] [Google Scholar]
  • 92.Loeb LA Human Cancers Express a Mutator Phenotype: Hypothesis, Origin, and Consequences. Cancer Res 76, 2057–2059 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Tomasetti C & Vogelstein B Cancer etiology. Variation in cancer risk among tissues can be explained by the number of stem cell divisions. Science 347, 78–81 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Popkin BM, Adair LS & Ng SW Global nutrition transition and the pandemic of obesity in developing countries. Nutr Rev 70, 3–21 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Stephen AM & Wald NJ Trends in individual consumption of dietary fat in the United States, 1920-1984. Am J Clin Nutr 52, 457–469 (1990). [DOI] [PubMed] [Google Scholar]
  • 96.Ng M et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: a systematic analysis for the Global Burden of Disease Study 2013. Lancet 384, 766–781 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.CDC. Achievements in Public Health, 1900-1999: Healthier Mothers and Babies. Morbidity and Mortality Weekly Report 48, 849–858 (1999). [PubMed] [Google Scholar]
  • 98.Simmons HE & Stolley PD This is medical progress? Trends and consequences of antibiotic use in the United States. JAMA 227, 1023–1028 (1974). [DOI] [PubMed] [Google Scholar]
  • 99.O'Sullivan A, Farver M & Smilowitz JT The Influence of Early Infant-Feeding Practices on the Intestinal Microbiome and Body Composition in Infants. Nutr Metab Insights 8, 1–9 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Lu YH, Wang N & Jin F Long-term follow-up of children conceived through assisted reproductive technology. J Zhejiang Univ Sci B 14, 359–371 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Sandall J et al. Short-term and long-term effects of caesarean section on the health of women and children. Lancet 392, 1349–1357 (2018). [DOI] [PubMed] [Google Scholar]
  • 102.Zhou Q et al. Risk of Colorectal Cancer in Ulcerative Colitis Patients: A Systematic Review and Meta-Analysis. Gastroenterol Res Pract 2019, 5363261 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Ghione S et al. Dramatic Increase in Incidence of Ulcerative Colitis and Crohn's Disease (1988-2011): A Population-Based Study of French Adolescents. Am J Gastroenterol 113, 265–272 (2018). [DOI] [PubMed] [Google Scholar]
  • 104.Mayer-Davis EJ et al. Incidence Trends of Type 1 and Type 2 Diabetes among Youths, 2002-2012. N Engl J Med 376, 1419–1429 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Poon SL, McPherson JR, Tan P, Teh BT & Rozen SG Mutation signatures of carcinogen exposure: genome-wide detection and new opportunities for cancer prevention. Genome Med 6, 24 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ogino S, Nowak JA, Hamada T, Milner DA Jr. & Nishihara R Insights into Pathogenic Interactions Among Environment, Host, and Tumor at the Crossroads of Molecular Pathology and Epidemiology. Annu Rev Pathol 14, 83–103 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Hur K et al. Hypomethylation of long interspersed nuclear element-1 (LINE-1) leads to activation of proto-oncogenes in human colorectal cancer metastasis. Gut 63, 635–646 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Cho YH et al. LINE-1 hypomethylation is associated with radiation-induced genomic instability in industrial radiographers. Environ Mol Mutagen 60, 174–184 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Gogna P, O'Sullivan DE & King WD The effect of inflammation-related lifestyle exposures and interactions with gene variants on long interspersed nuclear element-1 DNA methylation. Epigenomics 10, 785–796 (2018). [DOI] [PubMed] [Google Scholar]
  • 110.Martin EM & Fry RC Environmental Influences on the Epigenome: Exposure- Associated DNA Methylation in Human Populations. Annu Rev Public Health 39, 309–333 (2018). [DOI] [PubMed] [Google Scholar]
  • 111.Yan HHN et al. Organoid cultures of early-onset colorectal cancers reveal distinct and rare genetic profiles. Gut Epub ahead of print (2020). [DOI] [PubMed] [Google Scholar]
  • 112.Wirbel J et al. Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat Med 25, 679–689 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Yachida S et al. Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat Med 25, 968–976 (2019). [DOI] [PubMed] [Google Scholar]
  • 114.Chen B, Du G, Guo J & Zhang Y Bugs, drugs, and cancer: can the microbiome be a potential therapeutic target for cancer management? Drug Discov Today 24, 1000–1009 (2019). [DOI] [PubMed] [Google Scholar]
  • 115.Mima K et al. Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut 65, 1973–1980 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Brennan CA & Garrett WS Fusobacterium nucleatum - symbiont, opportunist and oncobacterium. Nat Rev Microbiol 17, 156–166 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Pleguezuelos-Manzano C et al. Mutational signature in colorectal cancer caused by genotoxic pks(+) E. coli. Nature 580, 269–273 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Dejea CM et al. Microbiota organization is a distinct feature of proximal colorectal cancers. Proc Natl Acad Sci U S A 111, 18321–18326 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Flemer B et al. Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut 66, 633–643 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Phipps AI, Chan AT & Ogino S Anatomic subsite of primary colorectal cancer and subsequent risk and distribution of second cancers. Cancer 119, 3140–3147 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Mima K et al. Fusobacterium nucleatum and T Cells in Colorectal Carcinoma. JAMA Oncol 1, 653–661 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kosumi K et al. The Amount of Bifidobacterium Genus in Colorectal Carcinoma Tissue in Relation to Tumor Characteristics and Clinical Outcome. Am J Pathol 188, 2839–2852 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Kim JY et al. Different risk factors for advanced colorectal neoplasm in young adults. World J Gastroenterol 22, 3611–3620 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Blaser MJ & Dominguez-Bello MG The Human Microbiome before Birth. Cell Host Microbe 20, 558–560 (2016). [DOI] [PubMed] [Google Scholar]
  • 125.Esaiassen E, Fjalstad JW, Juvet LK, van den Anker JN & Klingenberg C Antibiotic exposure in neonates and early adverse outcomes: a systematic review and meta-analysis. J Antimicrob Chemother 72, 1858–1870 (2017). [DOI] [PubMed] [Google Scholar]
  • 126.Azad MB et al. Impact of maternal intrapartum antibiotics, method of birth and breastfeeding on gut microbiota during the first year of life: a prospective cohort study. Bjog 123, 983–993 (2016). [DOI] [PubMed] [Google Scholar]
  • 127.Klein EY et al. Global increase and geographic convergence in antibiotic consumption between 2000 and 2015. Proc Natl Acad Sci U S A 115, E3463–e3470 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Park JE, Jardine L, Gottgens B, Teichmann SA & Haniffa M Prenatal development of human immunity. Science 368, 600–603 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Cao Y et al. Long-term use of antibiotics and risk of colorectal adenoma. Gut 67, 672–678 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Zhang J et al. Oral antibiotic use and risk of colorectal cancer in the United Kingdom, 1989-2012: a matched case-control study. Gut 68, 1971–1978 (2019). [DOI] [PubMed] [Google Scholar]
  • 131.Kilkkinen A et al. Antibiotic use predicts an increased risk of cancer. Int J Cancer 123, 2152–2155 (2008). [DOI] [PubMed] [Google Scholar]
  • 132.Mauri G et al. Early-onset colorectal cancer in young individuals. Mol Oncol 13, 109–131 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Ganal-Vonarburg SC, Hornef MW & Macpherson AJ Microbial-host molecular exchange and its functional consequences in early mammalian life. Science 368, 604–607 (2020). [DOI] [PubMed] [Google Scholar]
  • 134.Abualkhair WH et al. Trends in Incidence of Early-Onset Colorectal Cancer in the United States Among Those Approaching Screening Age. JAMA Netw Open 3, e1920407 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Ladabaum U, Dominitz JA, Kahi C & Schoen RE Strategies for Colorectal Cancer Screening. Gastroenterology 158, 418–432 (2020). [DOI] [PubMed] [Google Scholar]
  • 136.Enwerem N et al. Systematic Review of Prevalence, Risk Factors, and Risk for Metachronous Advanced Neoplasia in Patients With Young-Onset Colorectal Adenoma. Clin Gastroenterol Hepatol Epub ahead of print (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Rex DK et al. Colorectal Cancer Screening: Recommendations for Physicians and Patients From the U.S. Multi-Society Task Force on Colorectal Cancer. Gastroenterology 153, 307–323 (2017). [DOI] [PubMed] [Google Scholar]
  • 138.Wolf AMD et al. Colorectal cancer screening for average-risk adults: 2018 guideline update from the American Cancer Society. CA Cancer J Clin 68, 250–281 (2018). [DOI] [PubMed] [Google Scholar]
  • 139.Fedewa SA, Siegel RL, Goding Sauer A, Bandi P & Jemal A Colorectal cancer screening patterns after the American Cancer Society's recommendation to initiate screening at age 45 years. Cancer 126, 1351–1353 (2020). [DOI] [PubMed] [Google Scholar]
  • 140.Preventive US Services Task Force et al. Screening for Colorectal Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 315, 2564–2575 (2016). [DOI] [PubMed] [Google Scholar]
  • 141.Peterse EFP et al. The impact of the rising colorectal cancer incidence in young adults on the optimal age to start screening: Microsimulation analysis I to inform the American Cancer Society colorectal cancer screening guideline. Cancer 124, 2964–2973 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Ogino S, Chan AT, Fuchs CS & Giovannucci E Molecular pathological epidemiology of colorectal neoplasia: an emerging transdisciplinary and interdisciplinary field. Gut 60, 397–411 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Hughes LAE, Simons C, van den Brandt PA, van Engeland M & Weijenberg MP Lifestyle, Diet, and Colorectal Cancer Risk According to (Epi)genetic Instability: Current Evidence and Future Directions of Molecular Pathological Epidemiology. Curr Colorectal Cancer Rep 13, 455–469 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Gunter MJ et al. Meeting report from the joint IARC-NCI international cancer seminar series: a focus on colorectal cancer. Ann Oncol 30, 510–519 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Carr PR et al. Lifestyle factors and risk of sporadic colorectal cancer by microsatellite instability status: a systematic review and meta-analyses. Ann Oncol 29, 825–834 (2018). [DOI] [PubMed] [Google Scholar]
  • 146.Inamura K Roles of microbiota in response to cancer immunotherapy. Semin Cancer Biol Epub ahead of print (2020). [DOI] [PubMed] [Google Scholar]
  • 147.Rescigno T, Micolucci L, Tecce MF & Capasso A Bioactive Nutrients and Nutrigenomics in Age-Related Diseases. Molecules 22 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Wang ST et al. Tea polyphenols and their chemopreventive and therapeutic effects on colorectal cancer. World J Gastroenterol 26, 562–597 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Amitay EL et al. Association of Aspirin and Nonsteroidal Anti-Inflammatory Drugs With Colorectal Cancer Risk by Molecular Subtypes. J Natl Cancer Inst 111, 475–483 (2019). [DOI] [PubMed] [Google Scholar]
  • 150.Luo K et al. Fusobacterium nucleatum, the communication with colorectal cancer. Biomed Pharmacother 116, 108988 (2019). [DOI] [PubMed] [Google Scholar]
  • 151.Murphy N, Jenab M & Gunter MJ Adiposity and gastrointestinal cancers: epidemiology, mechanisms and future directions. Nat Rev Gastroenterol Hepatol 15, 659–670 (2018). [DOI] [PubMed] [Google Scholar]
  • 152.Rajpoot M, Sharma AK, Sharma A & Gupta GK Understanding the microbiome: Emerging biomarkers for exploiting the microbiota for personalized medicine against cancer. Semin Cancer Biol 52, 1–8 (2018). [DOI] [PubMed] [Google Scholar]
  • 153.Chavarro JE et al. Contributions of the Nurses' Health Studies to Reproductive Health Research. Am J Public Health 106, 1669–1676 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Gupta S et al. Potential impact of family history-based screening guidelines on the detection of early-onset colorectal cancer. Cancer Epub ahead of print (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Cirillo PM & Cohn BA Pregnancy complications and cardiovascular disease death: 50-year follow-up of the Child Health and Development Studies pregnancy cohort. Circulation 132, 1234–1242 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Dwyer AJ et al. A Summary of the Fight Colorectal Cancer Working Meeting: Exploring Risk Factors and Etiology of Sporadic Early-Age Onset Colorectal Cancer. Gastroenterology 157, 280–288 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Morton SM et al. Cohort profile: growing up in New Zealand. Int J Epidemiol 42, 65–75 (2013). [DOI] [PubMed] [Google Scholar]
  • 158.Connelly R & Platt L Cohort profile: UK Millennium Cohort Study (MCS). Int J Epidemiol 43, 1719–1725 (2014). [DOI] [PubMed] [Google Scholar]
  • 159.Nishi A et al. Lifecourse epidemiology and molecular pathological epidemiology. Am J Prev Med 48, 116–119 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Kim NH et al. Prevalence of and Risk Factors for Colorectal Neoplasia in Asymptomatic Young Adults (20-39 Years Old). Clin Gastroenterol Hepatol 17, 115–122 (2019). [DOI] [PubMed] [Google Scholar]

RESOURCES