Skip to main content
Clinical and Translational Gastroenterology logoLink to Clinical and Translational Gastroenterology
. 2025 May 28;16(8):e00864. doi: 10.14309/ctg.0000000000000864

The Enteric Microbiome in Early-Onset Colorectal Cancer: A Comprehensive Review of Its Role as a Biomarker of Disease

Jesús M Luévano Jr 1,, Julia Liu 1, Thaddeus Stappenbeck 2
PMCID: PMC12377310  PMID: 40434363

Abstract

Early-onset colorectal cancer (EoCRC), a distinct entity from late-onset colorectal cancer (LoCRC), continues to increase in incidence. Known risk factors for LoCRC have been explored to explain this trend, but do not account for it completely. The gastrointestinal microbiome has been associated with LoCRC and additional risk factors of disease; however, it is only now being investigated in the context of EoCRC. A better understanding of the microbiome's function in EoCRC could elucidate its role in the increasing incidence of EoCRC. This article reviews the state of literature related to studies specifically isolating microbiome-related changes in EoCRC compared with LoCRC and age-matched controls. Several studies reviewed in this article highlight the varied results of overall diversity and specific bacteria that are influenced by EoCRC, and the utility of these unique changes to predict for EoCRC. Although the microbiome can be useful in understanding EoCRC, to better predict for disease the microbiome must be studied in more diverse populations and with deeper, more functional characterization in a manner that allows for transference of findings among future studies. These studies indicate that the enteric microbiome holds significant potential as a biomarker for disease but has yet to fully meet an understanding necessary for direct clinical utilization.

KEYWORDS: microbiome, early-onset colorectal cancer, biomarker, review


graphic file with name ct9-16-e00864-g001.jpg

RISE OF EARLY-ONSET COLORECTAL CANCER

Colorectal cancer (CRC) ranks third in cancer mortality in the United States (1). Incidence increases with age, doubling in each successive 5-year group from ages 20 to 60 years (2). Improvements in mortality stem from universal screening, advanced surgical options, and newer adjuvant chemotherapies (3). However, early-onset colorectal cancer (EoCRC), defined as CRC before age 50 years, is rising in the United States, Europe, and Asia, accounting for 13% of all CRC, up from 6% 2 decades prior (1,49). Following updated specialty organizations recommendations (1012), in 2021, the US Preventive Services Task Force reduced the universal screening age to 45 years (13).

Most EoCRC cases seem sporadic, independent of genetic syndromes, with ∼70%–80% arising from somatic mutations (4,14,15). Compared with traditional CRC, dubbed average-onset CRC or late-onset CRC (LoCRC), EoCRC generally is more aggressive, diagnosed at advanced stages, and with greater metastatic potential (16,17). EoCRC incidence and mortality in Non-Hispanic Black-Americans is nearly double that of White patients (1820). Demographic factors associated with CRC also associate with EoCRC, including obesity and sedentary lifestyle (21,22), diabetes (23), and unhealthy diets (24,25). Of these, no singular factor provides a compelling explanation for the trend in EoCRC (7), with some studies demonstrating null results (26). Over the past 50 years, EoCRC incidence initially decreased from 1975 to 1990, but has since increased with differing magnitudes across 5-year age groups, suggestive of birth cohort effects in those born after 1960, possibly from early-life exposures (5,6,12,27).

Diet has well-demonstrated CRC associations, particularly Western diets low in fiber and rich in red meat (26,28,29) (vs prudent diets (30)), alcohol (24,26,28,31), and sugary drinks (32). Studies demonstrate stronger associations with Western diets and early-onset advanced adenomas in the distal colon/rectum (where EoCRC is more common) compared with the proximal colon (24), with high-fat avoidance being protective (25). Sugar-sweetened drinks associated with increased EoCRC risk (33), particularly in women (34), although fructose associated with increased proximal but not distal CRC mortality (35). Heavy alcohol use was associated with higher rates of EoCRC (3638), although 1 study found increased risk with abstinence and heavy use (39).

As further research links CRC risk factors to EoCRC, an important mechanism intertwined with metabolic syndromes, alcohol use, and dietary changes is the gastrointestinal microbiome (4047).

UPPER AND LOWER GASTROINTESTINAL MICROBIOME IN CRC

The human microbiome includes the totality of genomes encoding the transcriptional, translational, and metabolic functions of our commensal microbes. CRC-associated microbiome alterations, including decreased community diversity and shifts at higher-order taxonomic levels (e.g., Bacteroides and Firmicutes), have been sought for diagnostic, prophylactic, and therapeutic applications (4047). Patients with CRC had higher levels of Fusobacterium (42,4548), particularly mucosal samples (43), which produces the short-chain fatty acid (SCFA) butyrate, the primary colonocyte energy source and modulation of which is associated with CRC carcinogenesis (4951). ApcMin/+ mice exposed to F. nucleatum demonstrated increased tumor multiplicity and tumor-infiltrating myeloid cells, which can promote tumor progression (46). Interestingly, Enterobacteriaceae, which can produce DNA-damaging genotoxins (52), was decreased in CRC (45).

Common fecal bacteria associated with CRC carcinogenesis include Bacteroides fragilis (47,53), Escherichia coli (54), Enterococcus faecalis (55), Streptococcus gallolyticus (56), and Morganella morganii (57). The microbiome as a CRC biomarker has been demonstrated (5866). Several prevalent oral bacteria, Peptostreptococcus (43) and F. nucleatum (6769), have been associated with CRC, and unique oral microbiome shifts were successfully used to develop predictive models (60,62). Chen et al, developed predictive models for adenomas and CRC (area under the curve [AUC] = 0.84 and 0.93) using bacterial community and serum metabolomic data (59). Noninvasive screening alternatives featuring microbiome and metabolic data can improve upon accuracy of older serologic markers, cell-free DNA (7072), and even fecal-based immunochemical tests (73,74), a means to increase access for underserved populations (18,7577). However, few studies evaluate the role of the microbiome in EoCRC vs age matched controls or LoCRC.

MICROBIOME IN EoCRC

Sixteen studies were identified through searches on PubMed and Google Scholar using search items: “early onset colorectal cancer,” “EoCRC,” “microbiome,” “microbiota,” “16S,” “metagenomics,” “sequencing,” “metabolome,” and “young onset colorectal cancer.” Eleven were from the United States (7888) and 5 China (40,8992). All featured comparisons with EoCRC, varying in methodology for sample collection, sequencing, statistical analyses, and comparator group (LoCRC or age-matched controls). Prior studies primarily featured LoCRC or lacked age stratification (4048,5866). Only a few studies (40,81,8386,8891) and abstracts (7880,82,87) specifically evaluate microbiome alterations in EoCRC (Table 1).

Table 1.

Study design and patient metadata in early-onset colorectal cancer microbiome studies

Study Year Total subjects LoCRC cases (median age) EoCRC cases (median age) Controls (median age) Race: % (n) White/Black Sample types (sequencing methods) Study location PMID/DOI
Keshinro et ala,b 2020 275 114 (70)c 24 (33.6)c 137 Not stated Tumor tissue (NGS panel) New York, NY, USA 10.1200/jco.2020.38.15_suppl.e16070
Jin et ala 2022 358 304 54 0 Not stated Tumor tissue (RNA-Seq) Columbus, OH, USA 10.1016/j.annonc.2022.04.426
Kharofa et al 2022 1,301 611 81 609 (60) Not stated Stool (metagenome), meta-analysis of 11 studies Cincinnati, OH, USA 36056757
Dave et ala 2023 227 Not stated Not stated 0 Not stated Tumor tissue (16S) Cleveland, OH, USA 10.1200/jco.2023.41.4_suppl.13
Weinberg et al 2023 63 27 (72) 36 (38) 0 Not stated Tumor tissue (16S) Washington DC, USA 10.1200/jco.2023.41.16_suppl.3530
White et al 2023 107 70 (61) 37 (42) 0 71%/7.5% Tumor tissue and adjacent mucosa (metagenome) Houston, TX, USA 37465976
Adnan et al 2024 1,393 619 82 Age matched: EoCRC controls 125/LoCRC controls 568 Not stated Stool and mucosal tissue (16S) Chicago, IL, USA 37967575
Barot et al 2024 276 140 (43) 136 (73) 0 83.6% (117)/14.3% (20) Tumor tissue and adjacent mucosa (16S) Cleveland Clinic, OH, USA 38306898
Hong et ala 2024 377 0 377 0 Not stated Stool and mucosal tissue (16S, WGS, metabolomics) St. Louis, MO, USA 10.1200/jco.2024.42.16_suppl.e15646
Jayakrishnan et al 2024 64 44 20 0 100%/0% Tumor tissue (16S), serum (metabolomics) Cleveland Clinic, OH, USA 39020083
Jin et ala 2024 Not stated Not stated Not stated 0 Not stated Tumor tissue (RNA-Seq) Columbus, OH, USA 10.1200/jco.2024.42.16_suppl.3580
Yang et al 2021 1,038 379 (64) 185 (40) Age matched: EoCRC 217 controls (40)/LoCRC controls 257 (63) Not stated All stool (16S); stool subset of 200 samples (metagenome) Shanghai, China 34799562
Xiong et al 2022 98 43 24 31 Not stated Stool (16S) Harbin, China 36389150
Xu et al 2022 39 19 20 0 Not stated Tumor tissue (16S) Shanghai, China 36119074
Kong et al 2023 434 130 (62) 114 (40) Age matched: EoCRC controls 100 (39)/LoCRC controls 97 (62) Not stated Stool (metagenome/metabolomics) Shanghai, China 35953094
Qin et al 2024 460 293 167 0 Not stated Stool (metagenome), data from Yang et al and 8 public cohorts Guangzhou, China 38649355

Studies were separated based on country of origin (United States or China). Total subjects (by case status [late onset vs early onset] and controls) are presented, as well as median age for that groupc (except for Keshinro et al, which provided mean age). Early-onset cases were defined as cases diagnosed before age 50, except for Kenshinro et al, that defined them as diagnosed before age 40. Late-onset cases are defined as diagnosed after age 50. When available, racial demographics were included.

16S, 16S rRNA sequencing; EoCRC, early-onset colorectal cancer; LoCRC, late-onset colorectal cancer; NGS, next-generation sequencing; RNA-Seq, RNA sequencing; WGS, whole genome sequencing.

a

Abstracts.

b

Defined EoCRC as younger than 40 years.

c

Mean age.

Differences in EoCRC vs LoCRC

Several microbial differences were consistent between EoCRC and LoCRC. Within the phyla Fusobacteria, EoCRC was associated with increased abundance of Fusobacterium (40,88) and decreased Clostridium (83,88) and the family Leptotrichiaceae (83,85) (Figure 1).

Figure 1.

Figure 1.

Enteric microbiota alterations between EoCRC and LoCRC. Reported alterations of the enteric microbiota that featured comparisons of EoCRC vs LoCRC were summarized. Blue and red boxes, respectively, indicate reported increases and decreases in relative abundance associated with EoCRC. Bracketed letter indicates taxonomic level of classification (e.g., [p] indicates phylum). When available, species data are included within the genus taxa box. A summary of alpha diversity findings is included at the bottom of the table for each study. Bacterial taxa were grouped based on similar phyla or kingdom for fungi. One species, L. hofstadii, was seen only in patients with LoCRC. The samples from Adnan et al, comprised tumor-based samples only. EoCRC, early-onset colorectal cancer; LoCRC, late-onset colorectal cancer.

Greater proportions of Fusobacterium, associated with LoCRC in humans (42,4548,6769) and carcinogenesis in mice (43,46), provides a potential explanation for earlier and more aggressive disease in EoCRC. Clostridium decreased in CRC was increased after curative surgery, with alterations of carcinogenesis-associated deoxycholate bile acid (BA) producing function through bai operon, a microbiome-mediated functional change (93,94). C. septicum infections have been associated with CRC (95), and it grows well in the cecum (the most acidic colon segment (96)), whereas EoCRC more often presents distally, which may be related to lower proportions of Clostridium.

Within the phyla Pseudomonadota, decreases in Eschericia (83,86,87) and Pseudomonas (83,86) associated with EoCRC (Figure 1). Pseudomonas species can produce a biosurfactant with antitumorigenic properties against colon cancer cells (97), specifically the peptide Azurin-p28 that can enhance the effect of 5-fluorouracil (98). Loss of this capability may select for procarcinogenic states seen in EoCRC and explain why certain patients have greater 5-fluorouracil complications (99).

Remaining significant microbial taxa were limited to individual studies (Figure 1). There were several consistent bacterial associations with any CRC. Clostridium symbiosum was elevated in LoCRC and EoCRC compared with controls, as well as Peptostreptococcus stomatis, Parvimonas micra, and Hungatella hathewayi (92). This suggests predictive tools developed for LoCRC using microbial data may be able to successfully classify EoCRC. Alpha diversity variations in EoCRC, a measure of intraindividual microbial diversity, were not consistent, making it difficult to highlight a predictive signal.

Addressing the absence of global community shifts as a discriminatory signal will require deeper levels of microbial community profiling, larger training datasets, inclusion of other sampling sites, and functional profiling for better characterization.

Differences in EoCRC vs age-matched controls

Comparing EoCRC with age-matched controls found more consistent differences. Fusobacterium again was increased in EoCRC cases (40,87,89), except for 1 study (91), suggestive that EoCRC arises from luminal environments more procarcinogenic than LoCRC, especially compared with those without cancer (Figure 2).

Figure 2.

Figure 2.

Enteric microbiota alterations between EoCRC and age-matched controls. Reported alterations of the enteric microbiota that featured comparisons of EoCRC vs age-matched controls were summarized. Blue and red boxes, respectively, indicate reported increases and decreases in relative abundance associated with EoCRC or gray if mixed. Bracketed letter indicates taxonomic level of classification (e.g., [p] indicates phylum). When available, species data are included within the genus taxa box. A summary of alpha diversity findings is included at the bottom of the table for each study. Bacterial taxa were grouped based on similar phyla, and there were Archaea noted as well. In the study by Hong et al, the increased abundance of B. fragilis in EoCRC was found only in comparisons looking at tumor-adjacent tissue. EoCRC, early-onset colorectal cancer.

Several genera from the phyla Bacteroidetes were increased in EoCRC, including Porphyromonas (40,91) (specifically P. asaccharolytica (91)) and Bacteroides (84,85,87,89,91). B. fragilis was increased in EoCRC in several studies (85,87,91), concordant with prior findings in CRC (47,53) (Figure 2). Enterotoxigenic B. Fragilis produces fragilysin that can activate Wnt/B-catenin and nuclear factor kappa B pathways, inducing cell proliferation and inflammation which may trigger carcinogenic/inflammatory cascades in colonocytes to trigger myeloid cell–dependent distal colon tumorigenesis (100,101). Porphyromonas, and specifically P. asaccharolytica (63), has been well associated with LoCRC (42,43,63,102), another commonality with EoCRC.

Phyla Firmicutes had mixed results. Clostridioides difficile was decreased in EoCRC (87,91), whereas C. symbiosum was increased (84,92). C. symbiosum requires further investigation into its role in CRC. C. difficile, a pathogenic noncommensal associated with LoCRC, being depleted in EoCRC suggests nosocomial and opportunistic infections, and their predisposing risk factors, are less common in younger individuals and may play a smaller role in EoCRC pathogenesis (103).

Other bacteria elevated in EoCRC include Parvimonas micra (84,92) and Flavonifractor plauti (85,89,91) (Figure 2). Recent work with LoCRC has elucidated associations with oral microbiota, including Parvimonas micra now associated with EoCRC (59,63,64,104,105). Flavonifractor plauti is inversely associated with flavonoid compound ingestion, possibly linking dietary medicated influences to risk reduction (106).

Bacterial taxa significantly decreased in EoCRC include Blautia (89,91) (particularly B. hansenii (91)) and Faecalibacterium prausnitzii (85,89,91). Blautia and Faecalibacterium are associated with right-sided CRC, consistent with EoCRC commonly originating in the distal colon (107). Collinsella aerofaciens was mixed (84,91). Finally, Escherichia was increased in 2 studies (84,87), similar to prior findings with CRC (54) (Figure 2).

Alpha diversity was consistently lower in EoCRC compared with age-matched controls (40,89,91,92). Because the overall trend was more consistent than comparing CRC subtypes, alpha diversity may fare better as a biomarker for EoCRC in certain age populations at risk for cancer rather than classifying CRC subtypes.

Unique microbiome signals in EoCRC are promising, but only by improved profiling of structural and functional changes during pathogenesis may we elucidate potential causative factors for the increasing incidence of EoCRC in specific age populations that could become targets for screening or intervention. This influence may be further coupled with the multitude of risk factors well-established with CRC, which may induce their carcinogenic effects through the microbiome (Figure 3).

Figure 3.

Figure 3.

EoCRC risk factors influence the enteric microbiome. Environmental and host factors independently associated with colorectal cancer risk may also influence the gastrointestinal microbiome. These represent potential interlinked associations that could mediate the increased risk of EoCRC in the age cohort populations born after 1960 and presents potential targets for further investigation. EoCRC, early-onset colorectal cancer. Created in BioRender. Luevano J (2025). https://BioRender.com/g32x299.

Microbiome as noninvasive screening biomarker

Four studies evaluated the enteric microbiome as a biomarker (88,89,91,92). From their Fudan cohort, Yang et al, developed a random forest classifier (RFC) identifying EoCRC vs controls (AUC = 0.8657 on training, AUC = 0.7952 on validation). A new model combining all cases and controls accurately classified EoCRC (AUC = 0.878) and LoCRC (AUC = 0.8667) (89). Subsequent work featuring additional data had more variable accuracy, from an AUC = 0.5629 (Guangzhou cohort) up to 0.9042 (meta-dataset trained, Guangzhou tested) (92). The meta-cohort featured 293 LoCRC and 167 EoCRC Guanzhou cases, 379 LoCRC and 185 EoCRC Fudan cases, and aggregated 72 EoCRC and 528 LoCRC cases from 8 studies. Larger, more diverse cohorts improved the accuracy of microbiome-based prediction tools.

Using a RFC, Kong et al, evaluated the discriminatory ability of multiomic markers (bacterial metagenomics, stool metabolomics, and/or gene expression) for EoCRC vs controls with good accuracy (91). Forty-nine species (AUC = 0.8828 on test dataset, 0.7734 on validation), 36 stool metabolites (0.8828, 0.7535), 59 KO-genes (0.8395, 0.7552), and 27 integrated markers (0.9102, 0.7847) were used as prediction features (91). Jayakrishnan et al, used a Data Integration Analysis for Biomarker Discovery using latent variable approaches for Omics studies (DIABLO) classifier to discriminate EoCRC from LoCRC, featuring 16S data (from cases, AUC = 0.61) and untargeted serum metabolomics (AUC = 0.98). Controls were successfully age stratified through serum metabolomics (AUC = 0.79) (88).

This illustrates that additional multiomic markers improve on present noninvasive prediction methods and highlights the importance of increasing the accuracy of future predictive models by testing and tuning on broader populations. Meta-analyses provide a means to draw further conclusions from available data and can help address lack of diverse representation (81,92). Kharofa et al, used publicly available metagenomic data from 10 studies to identify microbial associations with EoCRC (81). Although the studies did not explicitly subset for EoCRC, in aggregate they included sufficient EoCRC cases to identify significant associations. Qin et al, incorporated EoCRC and age matched controls from a new cohort, prior cohort, and 8 publicly available datasets to develop a predictive pipeline with high accuracy featuring multinational data (92).

The studies reviewed include 847 US and 510 Chinese EoCRC cases; however, they feature methodological differences in sample collection (tissue vs feces) and sequencing methods for data uploaded to public repositories. Overcoming these differences requires careful consideration in statistical analyses. Downstream standardization would allow for evaluation of the effects from differing computational methods. Future studies should aim to sample throughout the gastrointestinal tract (i.e., tissue, saliva, and colon/stool) as well as serum and follow standardized processing pipelines to optimize cross-cohort characterization.

Deeper sequencing coupled with larger cohorts would improve species and strain level characterization in EoCRC to better determine microbial biomarkers. Studies incorporating data from non-EoCRC focused studies can be used but would be limited to those with publicly available metadata to accurately stratify by age and still face similar challenges stemming from methodological differences.

FUTURE DIRECTIONS AND DISCUSSION

Studies have demonstrated potential pathways by which pathogenic bacteria alter the luminal microenvironment in a way that is conducive to the pathogenesis of CRC (41,46,48,52,54,55,57,108112). However, CRC carcinogenesis is a multistep process influenced by host, environmental, and microbial factors (Figure 3). One postulated causal factor is antibiotic exposure which has a mild association with EoCRC, but significance was lost after covariate adjustment (113). The true influence of early-life antibiotic use has yet to be characterized through longitudinal studies.

Microbiome-metabolic associations

Certain host risk factors are mediated via the microbiome through SCFA production and BA biotransformation. Specific bacterial enzymes allow for deconjugation of primary BAs, which promotes retention in the lumen through to the distal colon where other bacteria can use them as substrates, and deconjugated BAs can be further transformed into secondary BAs (94) (Figure 4). Extreme diet shifts can alter the microbiome in just 2 weeks, with measurable microbiome and metabolome alterations that increase saccharolytic fermentation and butyrogenesis while suppressing secondary BA synthesis (114). Blautia, found to be decreased in EoCRC (85,89,91), harbors 3α-hydroxysteroid dehydrogenase (HSDH) activity to metabolize BA (115), whereas B. Fragilis (85,87,91), increased in EoCRC, can harbor 7α-HSDH activity, demonstrating the nuances in BA metabolism (116). However, further work directly linking these functions to EoCRC risk is needed.

Figure 4.

Figure 4.

Microbial mediation of dietary factors. Dietary factors that affect CRC risk are highlighted in 3 major food groups (protein/red meat, fiber/resistant starch, and fat/lipids). Although direct microbial metabolism of proteins is not well elucidated, they can lead to the production of SCFAs as well as toxic compounds that influence colon health. Fiber/resistant starches can be metabolized by the microbiome to produce SCFAs, in particular butyrate, that has many beneficial impacts on colonocytes. Fats and lipids induce BA secretion, and these can be biotransformed into secondary BAs that have carcinogenic potential, which is compounded when butyrate is deficient. BA, bile acid; CRC, colorectal cancer; SCFA, short-chain fatty acid. Created in BioRender. Luevano J (2025). https://BioRender.com/mv0u0sr.

Dietary fiber increases stool mass, bowel transit, and bacterial fermentation of resistant starches into SCFAs, improving colonocyte turnover, and in vitro inducing apoptosis in CRC cells (117121). Increased stool mass and reduced transit time can lower colonocyte exposure to carcinogens through physical dilution and shorter exposure time (Figure 4).

Balanced protein, fat, and fiber intake improves butyrate production reducing CRC risk (50), whereas Western diets promote protein fermentation and BA deconjugation, which is proinflammatory and damaging to colonocytes (51). The carcinogenic ability of secondary BAs are potentiated by butyrate deficiency (50). Roseburia and Faecalibacterium that produce butyrate have decreased levels in CRC (50) and EoCRC (85,89,91). This is particularly of note as butyrate-resistant colonocytes may retain growth advantages and potentially form more aggressive cancers through altered Histone Deacetylase function and insulin sensitivity/glucose tolerance (122,123).

Although human studies have yet to delineate mechanistically how metabolites from dietary fat influence CRC, animal models found that high-fat diets stimulated BA secretion, causing epithelium regression, mucosal damage, and increasing CRC risk (124,125). Heavy protein intake has been hypothesized to produce carcinogens like heterocyclic amines (HCA) and polycyclic aromatic hydrocarbons during excessive cooking, with HCA being highly mutagenic in vitro (126128). Protein fermentation differs from saccharolytic fermentation in that it produces many similar SCFAs from a carbon skeleton, but also releases potentially toxic nitrogenous and sulfur metabolites such as ammonia, amines, nitrates, nitrites, and hydrogen sulfide (129) (Figure 4).

By harnessing the microbiome, and better characterizing how known CRC risk factors mediate their influence through the microbiome in EoCRC, we may be able to not only predict for those at risk of EoCRC but reduce that risk (Figures 3 and 4).

Addressing EoCRC in underrepresented populations

The microbiome is influenced by host factors associated with health care disparities and race, often left out of investigative analyses. Racial disparity exists in CRC diagnosis and treatment and is more pronounced in EoCRC. Improved mortality from screening seen in White Americans is not fully realized in minority populations (130). Cumulative CRC survival and disease-specific mortality is worse for Black Americans compared with White patients across all disease stages, and they are up to 40% more likely to be diagnosed with advanced stage IV CRC (18,7577,131). Compared with White patients, Black patients had higher toxicity from 5-fluorouracil, lower rates of surgical resection, and shorter surveillance follow-up (99). Although race associates with worse outcomes, ultimately it may be an indirect marker of social and environmental risks that require improved comprehension (20,132135).

Pilot studies have investigated racial differences or focused on CRC in Black populations (136141). Comparing Blacks with other groups found differences that may influence disparities in CRC incidence and outcomes, especially considering differing microbiome baselines associated with host diversity (136,138,142,143). The gap is more pronounced in EoCRC studies. The majority lack racial or ethnic descriptions of their populations (40,7881,83,85,87,8992). Three include race descriptions, but feature predominantly White patients (71%–100%) (84,86,88). One abstract discussed differential abundance of taxa by race: with Black patients enriched for Limosilactobacillus, Bacillus, and Staphylococcus, whereas White patients were enriched for Enterococcus and Escherichia-Shigella (82). The microbiome of EoCRC and LoCRC cases were more similar within Black patients than EoCRC between races; however, population breakdown was not detailed (82). Currently we cannot conclude the influence and importance of race on EoCRC (Figure 5).

Figure 5.

Figure 5.

Key considerations to guide future work. Key concepts to improve further characterization of the role of the enteric microbiome in EoCRC and guide future investigative efforts summarized by focus categories. EoCRC, early-onset colorectal cancer.

Equalizing screening is a successful means to improve health care outcomes. The Delaware Cancer Consortium pioneered a program with navigators for CRC screening, cancer treatment for uninsured, and an emphasis on Black-American cancer reduction. Targeted efforts improved screening for Black patients from 48% in 2001 to 74% 2009, equal to rates in White patients (58%–74%) (144). By the study's end, there was no difference in LoCRC incidence, with mortality improvements lagging but nearly equalizing (144). However, not all health care systems can massively increase colonoscopies. Kaiser Permanente studied noninvasive screening as a method to eliminate disparities and found that proactive annual fecal immunochemical testing and on-request colonoscopy decreased absolute differences between Black and White patients from 21.6 to 1.6 cases per 100,000 (145).

Nonendoscopic screening alternatives based on microbiome and metabolic data demonstrate improved accuracy compared with older serologic markers (e.g., carcinoembryonic antigen and carbohydrate antigen 19-9) (7072,146148) or fecal-based immunochemical tests (73,74,149), supporting their diagnostic utility. Recently blood-based cell-free DNA screening was studied in average-risk populations (150), but additional investigation is needed to highlight at-risk and minority groups and build on the demonstrated utility of not only stool metabolite profiling but urine (151,152) and serum (88,152). Future studies should sample various noninvasive sources to better evaluate alterations associated with EoCRC carcinogenesis (Figure 5).

Improved comprehension is required into how demographic differences in vulnerable populations—such as minorities and those with EoCRC—are influencing the enteric microbiome and in turn CRC pathogenesis. Optimally quantifying these influences will require studies that incorporate community level analyses, as well as functional profiling and well-curated demographic information (e.g., dietary and medication information), to develop robust multiomic evaluation. Future tools would benefit from diverse training datasets to optimize development of new predictive tools with consistent accuracy when externally validated (Figure 5).

CONCLUSION

Our understanding of microbial alterations in patients with EoCRC compared with LoCRC and controls is improving but requires broader sampling in future studies. Evaluation of the microbiome along the gastrointestinal tract, characterization of functional alterations, and inclusion of diverse populations disproportionately burdened with disease will yield further insight into EoCRC. Future studies should capitalize on patient cohorts that serve underrepresented populations to better characterize how their microbiomes influence EoCRC, which may lead to worse outcomes, and develop broadly applicable prediction tools that could in turn be used to expand screening outreach and improve outcomes.

CONFLICTS OF INTEREST

Guarantor of the article: Jesús M. Luévano Jr, MD, MS.

Specific author contributions: J.M.L.: conceptualization and analysis of literature, and figure generation and writing-original draft preparation. J.L. and T.S.: supervision and editing. All authors have read and agreed to the published version of the manuscript.

Financial support: This work was supported by the American Cancer Society Diversity in Cancer Research Program—Clinician Scientist Development Grant (DICRIDG-21-072-01).

Potential competing interests: None to report.

ABBREVIATIONS:

16S

16S rRNA sequencing

aoCRC

average-onset colorectal cancer

AUC

area under the curve

BA

bile acid

CA 19-9

carbohydrate antigen 19-9

CEA

carcinoembryonic antigen

CRC

colorectal cancer

DIABLO

Data Integration Analysis for Biomarker Discovery using Latent variable approaches for Omics studies

EoCRC

early-onset colorectal cancer

HCA

heterocyclic amines

HSDH

hydroxysteroid dehydrogenase

LoCRC

late-onset colorectal cancer

NGS

next-generation sequencing

RFC

random forest classifier

RNA-Seq

RNA sequencing

SCFA

short-chain fatty acid

WGS

whole genome sequencing

Contributor Information

Julia Liu, Email: jjliu@msm.edu.

Thaddeus Stappenbeck, Email: STAPPET@ccf.org.

REFERENCES

  • 1.Siegel RL, Miller KD, Wagle NS, et al. Cancer statistics, 2023. CA Cancer J Clin 2023;73(1):17–48. [DOI] [PubMed] [Google Scholar]
  • 2.Ahnen DJ, Wade SW, Jones WF, et al. The increasing incidence of young-onset colorectal cancer: A call to action. Mayo Clin Proc 2014;89(2):216–24. [DOI] [PubMed] [Google Scholar]
  • 3.Kumar A, Gautam V, Sandhu A, et al. Current and emerging therapeutic approaches for colorectal cancer: A comprehensive review. World J Gastrointest Surg 2023;15(4):495–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Chang DT, Pai RK, Rybicki LA, et al. Clinicopathologic and molecular features of sporadic early-onset colorectal adenocarcinoma: An adenocarcinoma with frequent signet ring cell differentiation, rectal and sigmoid involvement, and adverse morphologic features. Mod Pathol 2012;25(8):1128–39. [DOI] [PubMed] [Google Scholar]
  • 5.Brim H, Reddy CS, Chirumamilla L, et al. Trends and symptoms among increasing proportion of African Americans with early-onset colorectal cancer over a 60-year period. Dig Dis Sci 2025;70(1):168–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Dwyer AJ, Murphy CC, Boland CR, et al. A summary of the fight colorectal cancer working meeting: Exploring risk factors and etiology of sporadic early-age onset colorectal cancer. Gastroenterology 2019;157(2):280–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Cavestro GM, Mannucci A, Zuppardo RA, et al. Early onset sporadic colorectal cancer: Worrisome trends and oncogenic features. Dig Liver Dis 2018;50(6):521–32. [DOI] [PubMed] [Google Scholar]
  • 8.Siegel RL, Miller KD, Fedewa SA, et al. Colorectal cancer statistics, 2017. CA Cancer J Clin 2017;67(3):177–93. [DOI] [PubMed] [Google Scholar]
  • 9.Venugopal A, Carethers JM. Epidemiology and biology of early onset colorectal cancer. EXCLI J 2022;21:162–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Issaka RB, Chan AT, Gupta S. AGA clinical practice update on risk stratification for colorectal cancer screening and post-polypectomy surveillance: Expert review. Gastroenterology 2023;165(5):1280–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Shaukat A, Kahi CJ, Burke CA, et al. ACG clinical guidelines: Colorectal cancer screening 2021. Am J Gastroenterol 2021;116(3):458–79. [DOI] [PubMed] [Google Scholar]
  • 12.Wolf AMD, Fontham ETH, Church TR, et al. Colorectal cancer screening for average-risk adults: 2018 guideline update from the American cancer society. CA Cancer J Clin 2018;68(4):250–81. [DOI] [PubMed] [Google Scholar]
  • 13.Davidson KW, Barry MJ, Mangione CM, et al. Screening for colorectal cancer: US Preventive Services Task Force recommendation statement. JAMA 2021;325(19):1965–77. [DOI] [PubMed] [Google Scholar]
  • 14.Strum WB, Boland CR. Clinical and genetic characteristics of colorectal cancer in persons under 50 years of age: A review. Dig Dis Sci 2019;64(11):3059–65. [DOI] [PubMed] [Google Scholar]
  • 15.Pearlman R, Frankel WL, Swanson B, et al. Prevalence and spectrum of germline cancer susceptibility gene mutations among patients with early-onset colorectal cancer. JAMA Oncol 2017;3(4):464–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Silla IO, Rueda D, Rodríguez Y, et al. Early-onset colorectal cancer: A separate subset of colorectal cancer. World J Gastroenterol 2014;20(46):17288–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Willauer AN, Liu Y, Pereira AAL, et al. Clinical and molecular characterization of early-onset colorectal cancer. Cancer 2019;125(12):2002–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.McClelland PHT, Liu T, Ozuner G. Early-onset colorectal cancer in patients under 50 years of age: Demographics, disease characteristics, and survival. Clin Colorectal Cancer 2022;21(2):e135–44. [DOI] [PubMed] [Google Scholar]
  • 19.Theuer CP, Wagner JL, Taylor TH, et al. Racial and ethnic colorectal cancer patterns affect the cost-effectiveness of colorectal cancer screening in the United States. Gastroenterology 2001;120(4):848–56. [DOI] [PubMed] [Google Scholar]
  • 20.Zaki TA, Liang PS, May FP, et al. Racial and ethnic disparities in early-onset colorectal cancer survival. Clin Gastroenterol Hepatol 2023;21(2):497–506.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Alvarez K, Cassana A, De La Fuente M, et al. Clinical, pathological and molecular characteristics of chilean patients with early-intermediate-and late-onset colorectal cancer. Cells 2021;10(3):631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Jin EH, Han K, Lee DH, et al. Association between metabolic syndrome and the risk of colorectal cancer diagnosed before age 50 years according to tumor location. Gastroenterology 2022;163(3):637–48.e2. [DOI] [PubMed] [Google Scholar]
  • 23.Khoa Ta HD, Nguyen NN, Ho DKN, et al. Association of diabetes mellitus with early-onset colorectal cancer: A systematic review and meta-analysis of 19 studies including 10 million individuals and 30,000 events. Diabetes Metab Syndr Clin Res Rev 2023;17(8):102828. [DOI] [PubMed] [Google Scholar]
  • 24.Huxley RR, Ansary-Moghaddam A, Clifton P, et al. The impact of dietary and lifestyle risk factors on risk of colorectal cancer: A quantitative overview of the epidemiological evidence. Int J Cancer 2009;125(1):171–80. [DOI] [PubMed] [Google Scholar]
  • 25.Khan NA, Hussain M, Rahman AU, et al. Dietary practices, addictive behavior and bowel habits and risk of early onset colorectal cancer: A case control study. Asian Pac J Cancer Prev 2015;16(17):7967–73. [DOI] [PubMed] [Google Scholar]
  • 26.Rosato V, Bosetti C, Levi F, et al. Risk factors for young-onset colorectal cancer. Cancer Causes Control 2013;24(2):335–41. [DOI] [PubMed] [Google Scholar]
  • 27.Murphy CC, Singal AG, Baron JA, et al. Decrease in incidence of young-onset colorectal cancer before recent increase. Gastroenterology 2018;155(6):1716–9.e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Moghaddam AA, Woodward M, Huxley R. Obesity and risk of colorectal cancer: A meta-analysis of 31 studies with 70,000 events. Cancer Epidemiol Biomarkers Prev 2007;16(12):2533–47. [DOI] [PubMed] [Google Scholar]
  • 29.Zheng X, Hur J, Nguyen LH, et al. Comprehensive assessment of diet quality and risk of precursors of early-onset colorectal cancer. J Natl Cancer Inst 2021;113(5):543–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Mehta RS, Nishihara R, Cao Y, et al. Association of dietary patterns with risk of colorectal cancer subtypes classified by Fusobacterium nucleatum in tumor tissue. JAMA Oncol 2017;3(7):921–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ferrari P, Jenab M, Norat T, et al. Lifetime and baseline alcohol intake and risk of colon and rectal cancers in the European Prospective Investigation into Cancer and Nutrition (EPIC). Int J Cancer 2007;121(9):2065–72. [DOI] [PubMed] [Google Scholar]
  • 32.Li Y, Guo L, He K, et al. Consumption of sugar-sweetened beverages and fruit juice and human cancer: A systematic review and dose-response meta-analysis of observational studies. J Cancer 2021;12(10):3077–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chang VC, Cotterchio M, De P, et al. Risk factors for early-onset colorectal cancer: A population-based case–control study in Ontario, Canada. Cancer Causes Control 2021;32(10):1063–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hur J, Otegbeye E, Joh HK, et al. Sugar-sweetened beverage intake in adulthood and adolescence and risk of early-onset colorectal cancer among women. Gut 2021;70(12):2330–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Yuan C, Joh HK, Wang QL, et al. Sugar-sweetened beverage and sugar consumption and colorectal cancer incidence and mortality according to anatomic subsite. Am J Clin Nutr 2022;115(6):1481–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Rueda M, Robertson Y, Acott A, et al. Association of tobacco and alcohol use with earlier development of colorectal pathology: Should screening guidelines be modified to include these risk factors? Am J Surg 2012;204(6):963–8. [DOI] [PubMed] [Google Scholar]
  • 37.Chen X, Li H, Guo F, et al. Alcohol consumption, polygenic risk score, and early- and late-onset colorectal cancer risk. eClinicalMedicine 2022;49:101460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Jin EH, Han K, Shin CM, et al. Sex and tumor-site differences in the association of alcohol intake with the risk of early-onset colorectal cancer. J Clin Oncol 2023;41(22):3816–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Archambault AN, Lin Y, Jeon J, et al. Nongenetic determinants of risk for early-onset colorectal cancer. JNCI Cancer Spectr 2021;5(3):pkab029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Xiong H, Wang J, Chang Z, et al. Gut microbiota display alternative profiles in patients with early-onset colorectal cancer. Front Cell Infect Microbiol 2022;12:1036946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Flemer B, Lynch DB, Brown JMR, et al. Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut 2017;66(4):633–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Ahn J, Sinha R, Pei Z, et al. Human gut microbiome and risk for colorectal cancer. J Natl Cancer Inst 2013;105(24):1907–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Chen W, Liu F, Ling Z, et al. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS One 2012;7(6):e39743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Sobhani I, Tap J, Roudot-Thoraval F, et al. Microbial dysbiosis in colorectal cancer (CRC) patients. PLoS One 2011;6(1):e16393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Marchesi JR, Dutilh BE, Hall N, et al. Towards the human colorectal cancer microbiome. PLoS One 2011;6(5):e20447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kostic AD, Chun E, Robertson L, et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 2013;14(2):207–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hale VL, Jeraldo P, Chen J, et al. Distinct microbes, metabolites, and ecologies define the microbiome in deficient and proficient mismatch repair colorectal cancers. Genome Med 2018;10(1):78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Yu J, Chen Y, Fu X, et al. Invasive Fusobacterium nucleatum may play a role in the carcinogenesis of proximal colon cancer through the serrated neoplasia pathway. Int J Cancer 2016;139(6):1318–26. [DOI] [PubMed] [Google Scholar]
  • 49.Karim MR, Iqbal S, Mohammad S, et al. Butyrate's (a short-chain fatty acid) microbial synthesis, absorption, and preventive roles against colorectal and lung cancer. Arch Microbiol 2024;206(4):137. [DOI] [PubMed] [Google Scholar]
  • 50.Yang J, Yu J. The association of diet, gut microbiota and colorectal cancer: What we eat may imply what we get. Protein Cell 2018;9(5):474–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.O'Keefe SJD. Diet, microorganisms and their metabolites, and colon cancer. Nat Rev Gastroenterol Hepatol 2016;13(12):691–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Nougayrède JP Homburg S Taieb F, et al. Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science. 2006;313(5788):848–51. [DOI] [PubMed] [Google Scholar]
  • 53.Novielli P, Romano D, Magarelli M, et al. Explainable artificial intelligence for microbiome data analysis in colorectal cancer biomarker identification. Front Microbiol 2024;15:1348974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Arthur JC, Perez-Chanona E, Mühlbauer M, et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 2012;338(6103):120–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Huycke MM, Abrams V, Moore DR. Enterococcus faecalis produces extracellular superoxide and hydrogen peroxide that damages colonic epithelial cell DNA. Carcinogenesis. 2002;23(3):529–36. [DOI] [PubMed] [Google Scholar]
  • 56.Boleij A, Tjalsma H. The itinerary of Streptococcus gallolyticus infection in patients with colonic malignant disease. Lancet Infect Dis. 2013;13(8):719–24. [DOI] [PubMed] [Google Scholar]
  • 57.Cao Y, Oh J, Xue M, et al. Commensal microbiota from patients with inflammatory bowel disease produce genotoxic metabolites. Science 2022;378(6618):eabm3233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Zwezerijnen-Jiwa FH, Sivov H, Paizs P, et al. A systematic review of microbiome-derived biomarkers for early colorectal cancer detection. Neoplasia (United States) 2023;36:100868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Chen F, Dai X, Zhou CC, et al. Integrated analysis of the faecal metagenome and serum metabolome reveals the role of gut microbiome-associated metabolites in the detection of colorectal cancer and adenoma. Gut 2022;71(7):1315–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Zhang S, Kong C, Yang Y, et al. Human oral microbiome dysbiosis as a novel non-invasive biomarker in detection of colorectal cancer. Theranostics 2020;10(25):11595–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Wirbel J, Pyl PT, Kartal E, et al. Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat Med 2019;25(4):679–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Flemer B, Warren RD, Barrett MP, et al. The oral microbiota in colorectal cancer is distinctive and predictive. Gut 2018;67(8):1454–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Dai Z, Coker OO, Nakatsu G, et al. Multi-cohort analysis of colorectal cancer metagenome identified altered bacteria across populations and universal bacterial markers. Microbiome 2018;6(1):70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Yu J, Feng Q, Wong SH, et al. Metagenomic analysis of faecal microbiome as a tool towards targeted non-invasive biomarkers for colorectal cancer. Gut 2017;66(1):70–8. [DOI] [PubMed] [Google Scholar]
  • 65.Zackular JP, Rogers MAM, Ruffin MT, et al. The human gut microbiome as a screening tool for colorectal cancer. Cancer Prev Res (Phila) 2014;7(11):1112–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Zeller G, Tap J, Voigt AY, et al. Potential of fecal microbiota for early-stage detection of colorectal cancer. Mol Syst Biol 2014;10(11):766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Ou S Wang H Tao Y, et al. Fusobacterium nucleatum and colorectal cancer: From phenomenon to mechanism. Front Cell Infect Microbiol. 2022;12:1020583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Shimomura Y Sugi Y Kume A, et al. Strain-level detection of Fusobacterium nucleatum in colorectal cancer specimens by targeting the CRISPR-Cas region. Microbiol Spectr. 2023;11(6):e0512322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Zepeda-Rivera M Minot SS Bouzek H, et al. A distinct Fusobacterium nucleatum clade dominates the colorectal cancer niche. Nature. 2024;628(8007):424–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Li C, Zhao K, Zhang D, et al. Prediction models of colorectal cancer prognosis incorporating perioperative longitudinal serum tumor markers: A retrospective longitudinal cohort study. BMC Med 2023;21(1):63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Zhang Y, Wang Y, Zhang B, et al. Methods and biomarkers for early detection, prediction, and diagnosis of colorectal cancer. Biomed Pharmacother 2023;163:114786. [DOI] [PubMed] [Google Scholar]
  • 72.Lo YMD. Cell-free DNA for colorectal cancer screening. N Engl J Med 2024;390(11):1047–50. [DOI] [PubMed] [Google Scholar]
  • 73.Lee JK, Liles EG, Bent S, et al. Accuracy of fecal immunochemical tests for colorectal cancer: Systematic review and meta-analysis. Ann Intern Med 2014;160(3):171–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Imperiale TF, Ransohoff DF, Itzkowitz SH, et al. Multitarget stool DNA testing for colorectal-cancer screening. N Engl J Med 2014;370(14):1287–97. [DOI] [PubMed] [Google Scholar]
  • 75.Holowatyj AN, Ruterbusch JJ, Rozek LS, et al. Racial/ethnic disparities in survival among patients with young-onset colorectal cancer. J Clin Oncol 2016;34(18):2148–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Ollberding NJ, Nomura AMY, Wilkens LR, et al. Racial/ethnic differences in colorectal cancer risk: The multiethnic cohort study. Int J Cancer 2011;129(8):1899–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Primm KM, Malabay AJ, Curry T, et al. Who, where, when: Colorectal cancer disparities by race and ethnicity, subsite, and stage. Cancer Med 2023;12(13):14767–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Keshinro A, Vanderbilt C, Stadler ZK, et al. Do differences in the microbiome explain early onset in colon cancer? J Clin Oncol 2020;38(15 Suppl):e16070. [Google Scholar]
  • 79.Jin N, Yilmaz A, Hoyd R, et al. SO-27 Microbiome signature, global methylation and immune landscape in early onset colorectal cancer. Ann Oncol 2022;33:S368. [Google Scholar]
  • 80.Jin N, Hoyd R, Yilmaz AS, et al. Microbiome signature, immune landscape and global methylation in early onset colorectal cancer. J Clin Oncol 2024;42(16 Suppl):3580. [Google Scholar]
  • 81.Kharofa J, Apewokin S, Alenghat T, et al. Metagenomic analysis of the fecal microbiome in colorectal cancer patients compared to healthy controls as a function of age. Cancer Med 2023;12(3):2945–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Dave HM, Sangwan N, Nair KG, et al. Racial disparities in tumor microbiome in young-onset and average-onset colorectal cancer. J Clin Oncol 2023;41(4 Suppl):13. [Google Scholar]
  • 83.Weinberg BA, Wang H, Geng X, et al. Comprehensive study of the intratumoral microbiome in early- vs. late-onset colorectal cancer: Final analysis of COSMO CRC. J Clin Oncol 2023;41(16 Suppl):3530. [Google Scholar]
  • 84.White MG, Damania A, Alshenaifi J, et al. Young-onset rectal cancer: Unique tumoral microbiome and correlation with response to neoadjuvant therapy. Ann Surg 2023;278(4):538–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Adnan D, Trinh JQ, Sharma D, et al. Early-onset colon cancer shows a distinct intestinal microbiome and a host-microbe interaction. Cancer Prev Res 2024;17(1):29–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Barot SV, Sangwan N, Nair KG, et al. Distinct intratumoral microbiome of young-onset and average-onset colorectal cancer. eBioMedicine 2024;100:104980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hong D, Tian R, Kim H, et al. The microbiome landscape of early-onset colorectal cancer: A systematic review. J Clin Oncol 2024;42(16 Suppl):e15646. [Google Scholar]
  • 88.Jayakrishnan TT, Sangwan N, Barot SV, et al. Multi-omics machine learning to study host-microbiome interactions in early-onset colorectal cancer. npj Precis Oncol 2024;8(1):146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Yang Y, Du L, Shi D, et al. Dysbiosis of human gut microbiome in young-onset colorectal cancer. Nat Commun 2021;12(1):6757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Xu Z, Lv Z, Chen F, et al. Dysbiosis of human tumor microbiome and aberrant residence of Actinomyces in tumor-associated fibroblasts in young-onset colorectal cancer. Front Immunol 2022;13:1008975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kong C, Liang L, Liu G, et al. Integrated metagenomic and metabolomic analysis reveals distinct gut-microbiome-derived phenotypes in early-onset colorectal cancer. Gut 2023;72(6):1129–42. [DOI] [PubMed] [Google Scholar]
  • 92.Qin Y, Tong X, Mei WJ, et al. Consistent signatures in the human gut microbiome of old- and young-onset colorectal cancer. Nat Commun 2024;15(1):3396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Hiranyakas A, Ho YH. Surgical treatment for colorectal cancer. Int Surg 2011;96(2):120–6. [DOI] [PubMed] [Google Scholar]
  • 94.Fogelson KA, Dorrestein PC, Zarrinpar A, et al. The gut microbial bile acid modulation and its relevance to digestive health and diseases. Gastroenterology 2023;164(7):1069–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Larson CM, Bubrick MP, Jacobs DM, et al. Malignancy, mortality, and medicosurgical management of Clostridium septicum infection. Surgery 1995;118(4):592–8. [DOI] [PubMed] [Google Scholar]
  • 96.Fallingborg J. Intraluminal pH of the human gastrointestinal tract. Dan Med Bull 1999;46(3):183–96. [PubMed] [Google Scholar]
  • 97.Haque E, Hassan S. Physiochemical characterization and anti-colon cancer activity of biosurfactant produced from marine Pseudomonas sp. Int J Pharm Investig 2020;10(2):136–40. [Google Scholar]
  • 98.Yaghoubi A Movaqar A Asgharzadeh F, et al. Anticancer activity of Pseudomonas aeruginosa derived peptide with iRGD in colon cancer therapy. Iran J Basic Med Sci. 2023;26(7):768–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Dimou A, Syrigos KN, Saif MW. Disparities in colorectal cancer in African-Americans vs Whites: Before and after diagnosis. World J Gastroenterol 2009;15(30):3734–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Chung L, Orberg ET, Geis AL, et al. Carcinogenic inflammatory cascade via targeting. Cell Host Microbe 2018;23(2):203–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Shiryaev SA, Remacle AG, Chernov AV, et al. Substrate cleavage profiling suggests a distinct function of bacteroides fragilis metalloproteinases (fragilysin and metalloproteinase II) at the microbiome-inflammation-cancer interface. J Biol Chem 2013;288(48):34956–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Sinha R, Ahn J, Sampson JN, et al. Fecal microbiota, fecal metabolome, and colorectal cancer interrelations. PLoS One 2016;11(3):e0152126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Anderson SM, Sears CL. The role of the gut microbiome in cancer: A review, with special focus on colorectal neoplasia and Clostridioides difficile. Clin Infect Dis. 2023;77(Suppl 6):S471-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Drewes JL, White JR, Dejea CM, et al. High-resolution bacterial 16S rRNA gene profile meta-analysis and biofilm status reveal common colorectal cancer consortia. npj Biofilms Microbiomes 2017;3(1):34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Zhao L, Cho WC, Nicolls MR. Colorectal cancer-associated microbiome patterns and signatures. Front Genet 2021;12:787176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Ivey KL, Chan AT, Izard J, et al. Role of dietary flavonoid compounds in driving patterns of microbial community assembly. MBio 2019;10(5):e01205-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Miyake T, Mori H, Yasukawa D, et al. The comparison of fecal microbiota in left-side and right-side human colorectal cancer. Eur Surg Res 2021;62(4):248–54. [DOI] [PubMed] [Google Scholar]
  • 108.Moon JY, Kye BH, Ko SH, et al. Sulfur metabolism of the gut microbiome and colorectal cancer: The threat to the younger generation. Nutrients 2023;15(8):1966. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Wu S, Rhee KJ, Albesiano E, et al. A human colonic commensal promotes colon tumorigenesis via activation of T helper type 17 T cell responses. Nat Med 2009;15(9):1016–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Abdulamir AS, Hafidh RR, Bakar FA. Molecular detection, quantification, and isolation of Streptococcus gallolyticus bacteria colonizing colorectal tumors: Inflammation-driven potential of carcinogenesis via IL-1, COX-2, and IL-8. Mol Cancer. 2010;9:249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Arthur JC, Gharaibeh RZ, Mühlbauer M, et al. Microbial genomic analysis reveals the essential role of inflammation in bacteria-induced colorectal cancer. Nat Commun 2014;5:4724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Boleij A, Hechenbleikner EM, Goodwin AC, et al. The bacteroides fragilis toxin gene is prevalent in the colon mucosa of colorectal cancer patients. Clin Infect Dis 2015;60(2):208–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Kane KJ, Jensen CD, Yang J, et al. Oral antibiotic use in adulthood and risk of early-onset colorectal cancer: A case-control study. Clin Gastroenterol Hepatol 2024. doi: 10.1016/j.cgh.2024.09.002 [DOI] [PubMed] [Google Scholar]
  • 114.O'Keefe SJD, Li JV, Lahti L, et al. Fat, fibre and cancer risk in African Americans and rural Africans. Nat Commun 2015;6:6342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Devlin AS, Fischbach MA. A biosynthetic pathway for a prominent class of microbiota-derived bile acids. Nat Chem Biol 2015;11(9):685–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Li R, Andreu-Sánchez S, Kuipers F, et al. Gut microbiome and bile acids in obesity-related diseases. Best Pract Res Clin Endocrinol Metab 2021;35(3):101493. [DOI] [PubMed] [Google Scholar]
  • 117.Burkitt DP. Epidemiology of cancer of the colon and rectum. Dis Colon Rectum 1993;36(11):1071–82. [DOI] [PubMed] [Google Scholar]
  • 118.Hamer HM, Jonkers D, Venema K, et al. Review article: The role of butyrate on colonic function. Aliment Pharmacol Ther 2008;27(2):104–19. [DOI] [PubMed] [Google Scholar]
  • 119.Bergman EN. Energy contributions of volatile fatty acids from the gastrointestinal tract in various species. Physiol Rev 1990;70(2):567–90. [DOI] [PubMed] [Google Scholar]
  • 120.Fung KYC, Cosgrove L, Lockett T, et al. A review of the potential mechanisms for the lowering of colorectal oncogenesis by butyrate. Br J Nutr 2012;108(5):820–31. [DOI] [PubMed] [Google Scholar]
  • 121.Kim YH, Park JW, Lee JY, et al. Sodium butyrate sensitizes TRAIL-mediated apoptosis by induction of transcription from the DR5 gene promoter through Sp1 sites in colon cancer cells. Carcinogenesis 2004;25(10):1813–20. [DOI] [PubMed] [Google Scholar]
  • 122.Serpa J, Caiado F, Carvalho T, et al. Butyrate-rich colonic microenvironment is a relevant selection factor for metabolically adapted tumor cells. J Biol Chem 2010;285(50):39211–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.López de Silanes I, Olmo N, Turnay J, et al. Acquisition of resistance to butyrate enhances survival after stress and induces malignancy of human colon carcinoma cells. Cancer Res 2004;64(13):4593–600. [DOI] [PubMed] [Google Scholar]
  • 124.Narisawa T, Magadia NE, Weisburger JH, et al. Promoting effect of bile acids on colon carcinogenesis after intrarectal instillation of N-methyl-N′ nitro-N-nitrosoguanidine in rats. J Natl Cancer Inst 1974;53(4):1093–7. [DOI] [PubMed] [Google Scholar]
  • 125.Chomchai C, Bhadrachari N, Nigro ND. The effect of bile on the induction of experimental intestinal tumors in rats. Dis Colon Rectum 1974;17(3):310–2. [DOI] [PubMed] [Google Scholar]
  • 126.Nagao M. A new approach to risk estimation of food-borne carcinogens: Heterocyclic amines: Based on molecular information. Mutat Res 1999;431(1):3–12. [DOI] [PubMed] [Google Scholar]
  • 127.Nagao M, Ushijima T, Toyota M, et al. Genetic changes induced by heterocyclic amines. Mutat Res 1997;376(1-2):161–7. [DOI] [PubMed] [Google Scholar]
  • 128.Burnouf DY, Miturski R, Nagao M, et al. Early detection of 2-amino-1-methyl-6-phenylimidazo (4,5-b)pyridine(PhIP)-induced mutations within the Apc gene of rat colon. Carcinogenesis 2001;22(2):329–35. [DOI] [PubMed] [Google Scholar]
  • 129.Windey K, de Preter V, Verbeke K. Relevance of protein fermentation to gut health. Mol Nutr Food Res 2012;56(1):184–96. [DOI] [PubMed] [Google Scholar]
  • 130.Jackson CS, Oman M, Patel AM, et al. Health disparities in colorectal cancer among racial and ethnic minorities in the United States. J Gastrointest Oncol 2016;7(Suppl 1):S32–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Chien C, Morimoto LM, Tom J, et al. Differences in colorectal carcinoma stage and survival by race and ethnicity. Cancer 2005;104(3):629–39. [DOI] [PubMed] [Google Scholar]
  • 132.Kane WJ, Fleming MA, Lynch KT, et al. Associations of race, ethnicity, and social determinants of health with colorectal cancer screening. Dis Colon Rectum 2023;66(9):1223–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Mitsakos AT, Irish W, Parikh AA, et al. The association of health insurance and race with treatment and survival in patients with metastatic colorectal cancer. PLoS One 2022;17(2):e0263818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Obrochta CA, Murphy JD, Tsou MH, et al. Disentangling racial, ethnic, and socioeconomic disparities in treatment for colorectal cancer. Cancer Epidemiol Biomarkers Prev 2021;30(8):1546–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Tramontano AC, Chen Y, Watson TR, et al. Racial/ethnic disparities in colorectal cancer treatment utilization and phase-specific costs, 2000–2014. PLoS One 2020;15(4):e0231599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Yazici C, Wolf PG, Kim H, et al. Race-dependent association of sulfidogenic bacteria with colorectal cancer. Gut 2017;66(11):1983–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Carson TL, Wang F, Cui X, et al. Associations between race, perceived psychological stress, and the gut microbiota in a sample of generally healthy black and white women: A pilot study on the role of race and perceived psychological stress. Psychosom Med 2018;80(7):640–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Farhana L, Antaki F, Murshed F, et al. Gut microbiome profiling and colorectal cancer in African Americans and Caucasian Americans. World J Gastrointest Pathophysiol 2018;9(2):47–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Carson TL, Byrd DA, Smith KS, et al. A case–control study of the association between the gut microbiota and colorectal cancer: Exploring the roles of diet, stress, and race. Gut Pathog 2024;16(1):13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Hein DM, Coughlin LA, Poulides N, et al. Assessment of distinct gut microbiome signatures in a diverse cohort of patients undergoing definitive treatment for rectal cancer. J Immunother Precis Oncol 2024;7(3):150–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Piawah S, Kyaw TS, Trepka K, et al. Associations between the gut microbiota, race, and ethnicity of patients with colorectal cancer: A pilot and feasibility study. Cancers (Basel) 2023;15(18):4546. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Deschasaux M, Bouter KE, Prodan A, et al. Depicting the composition of gut microbiota in a population with varied ethnic origins but shared geography. Nat Med 2018;24(10):1526–31. [DOI] [PubMed] [Google Scholar]
  • 143.Brooks AW, Priya S, Blekhman R, et al. Gut microbiota diversity across ethnicities in the United States. PLoS Biol 2018;16(12):e2006842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Grubbs SS, Polite BN, Carney J, et al. Eliminating racial disparities in colorectal cancer in the real world: It took a village. J Clin Oncol 2013;31(16):1928–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Doubeni CA, Corley DA, Zhao W, et al. Association between improved colorectal screening and racial disparities. N Engl J Med 2022;386(8):796–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Guo X, Wang R, Chen R, et al. Gut microbiota and serum metabolite signatures along the colorectal adenoma-carcinoma sequence: Implications for early detection and intervention. Clin Chim Acta 2024;560:119732. [DOI] [PubMed] [Google Scholar]
  • 147.Xie Z, Zhu R, Huang X, et al. Metabolomic analysis of gut metabolites in patients with colorectal cancer: Association with disease development and outcome. Oncol Lett 2023;26(2):358. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Yagin FH, Alkhateeb A, Colak C, et al. A fecal-microbial-extracellular-vesicles-based metabolomics machine learning framework and biomarker discovery for predicting colorectal cancer patients. Metabolites 2023;13(5):589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Clos-Garcia M, Garcia K, Alonso C, et al. Integrative analysis of fecal metagenomics and metabolomics in colorectal cancer. Cancers (Basel) 2020;12(5):1142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Chung DC, Gray DM, Singh H, et al. A cell-free DNA blood-based test for colorectal cancer screening. N Engl J Med 2024;390(11):973–83. [DOI] [PubMed] [Google Scholar]
  • 151.Udo R, Katsumata K, Kuwabara H, et al. Urinary charged metabolite profiling of colorectal cancer using capillary electrophoresis-mass spectrometry. Sci Rep 2020;10(1):21057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Sun J, Zhao J, Zhou S, et al. Systematic investigation of genetically determined plasma and urinary metabolites to discover potential interventional targets for colorectal cancer. JNCI J Natl Cancer Inst 2024;116(8):1303–12. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical and Translational Gastroenterology are provided here courtesy of American College of Gastroenterology

RESOURCES