Skip to main content
Erschienen in: BMC Medicine 1/2022

Open Access 01.12.2022 | Research article

Short-chain fatty acid concentrations in the incidence and risk-stratification of colorectal cancer: a systematic review and meta-analysis

verfasst von: Ehsan Alvandi, Wilson K. M. Wong, Mugdha V. Joglekar, Kevin J. Spring, Anandwardhan A. Hardikar

Erschienen in: BMC Medicine | Ausgabe 1/2022

Abstract

Background

The beneficial role of gut microbiota and bacterial metabolites, including short-chain fatty acids (SCFAs), is well recognized, although the available literature around their role in colorectal cancer (CRC) has been inconsistent.

Methods

We performed a systematic review and meta-analysis to examine the associations of fecal SCFA concentrations to the incidence and risk of CRC. Data extraction through Medline, Embase, and Web of Science was carried out from database conception to June 29, 2022. Predefined inclusion/exclusion criteria led to the selection of 17 case-control and six cross-sectional studies for quality assessment and analyses. Studies were categorized for CRC risk or incidence, and RevMan 5.4 was used to perform the meta-analyses. Standardized mean differences (SMD) with 95% confidence intervals (CI) were calculated using a random-effects model. Studies lacking quantitation were included in qualitative analyses.

Results

Combined analysis of acetic, propionic, and butyric acid revealed significantly lower concentrations of these SCFAs in individuals with a high-risk of CRC (SMD = 2.02, 95% CI 0.31 to 3.74, P = 0.02). Additionally, CRC incidence was higher in individuals with lower levels of SCFAs (SMD = 0.45, 95% CI 0.19 to 0.72, P = 0.0009), compared to healthy individuals. Qualitative analyses identified 70.4% of studies reporting significantly lower concentrations of fecal acetic, propionic, butyric acid, or total SCFAs in those at higher risk of CRC, while 66.7% reported significantly lower concentrations of fecal acetic and butyric acid in CRC patients compared to healthy controls.

Conclusions

Overall, lower fecal concentrations of the three major SCFAs are associated with higher risk of CRC and incidence of CRC.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12916-022-02529-4.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CRC
Colorectal cancer
CRA
Colorectal adenoma
SCFA
Short-chain fatty acid
GLOBOCAN
Global Cancer Incidence, Mortality and Prevalence
PRISMA
Preferred Reporting Items for Systematic Reviews and Meta-Analyses
NOS
Newcastle-Ottawa Scale
JBI
Joanna Briggs Institute
RevMan
Review Manager
SMD
Standardized mean difference
RS
Resistance starch
IBS
Irritable bowel syndrome
IBD
Inflammatory bowel disease
EOCRC
Early-onset colorectal cancer
gFOBT
Guaiac fecal occult blood test
FIT
Fecal immunochemical test

Background

According to the Global Cancer Incidence, Mortality and Prevalence (GLOBOCAN) 2020 report, colorectal cancer (CRC) is the third most commonly diagnosed cancer (10% of all diagnosed cancers) and the second (9.4%) leading cause of cancer-related death [1]. It has been estimated that the overall risk of CRC in all age groups will increase 60% worldwide by 2030, leading to more than 1.1 million deaths and 2.2 million new cases [2]. Colorectal cancer develops from precursor lesions collectively known as colorectal adenomas (CRA), in the form of adenomatous polyps or to a lesser extent (10–20%) serrated polyps [3, 4]. It is a heterogeneous disease and environmental factors have a potential impact on the development of CRC, among which diet is a risk factor [46]. According to several meta-analyses, high consumption of processed and unprocessed meat is related to high CRC risk [7, 8], and high fiber intake is suggested as a protective factor against CRC progression and incidence [911].
The effect of diet on colonic health is partly mediated through alteration of gut microbiota composition, diversity, and metabolism [6, 12]. Gut microbiota constitutes the largest community of commensal microorganisms in the body, which mainly resides in the lower small intestine and colon [6, 12, 13]. The gut microbiota-derived metabolites are in constant crosstalk with colonocytes, and short-chain fatty acids (SCFAs) make up a large group of these metabolites [6, 12, 13].
Short-chain fatty acids are small molecules generated via the fermentation of dietary fibers by gut microbiota. Acetic, propionic, and butyric acid constitutes the majority of colonic SCFA content [14, 15] and the beneficial anti-inflammatory and anti-carcinogenic effects of dietary fibers on colonocytes are mediated through these SCFA molecules [16, 17]. Among the three major SCFA molecules, butyric acid is also considered as one of the main energy sources for colonocytes [12, 14, 17]. Therefore, alteration in SCFA levels could impact the colonic health and predisposition of colonocytes to aberrant proliferation and tumor formation [15, 16].
Several studies have assessed fecal SCFA concentration in patients with colorectal carcinoma or adenoma [1834]. However, due to variable results, the conclusive evaluation of SCFA profiles from CRC patients versus healthy subjects is lacking. In addition, other studies have compared SCFA concentration within healthy individuals from various countries and ethnic groups with the highest and lowest prevalence of CRC; although with inconsistent results [3540].
Therefore, systematic analyses designed to better understand the link between SCFA concentration in CRC risk and incidence is highly desired. We divided our analyses on the available evidence into two broad categories: (1) CRC-risk and (2) incidence. We aimed to systematically analyze the results of all primary observational human studies, which measured fecal SCFA levels in “at-risk” individuals or in CRC patients. In the CRC risk category, the focus was on at-risk individuals, which was further sub-divided into two groups based on (1a) studies that analyzed clinical data (presence of colorectal adenomas) or (1b) those that assigned CRC risk based on non-clinical evaluation of study participants (ethnic background or location). The CRC incidence category included studies that compared fecal SCFA levels in individuals with clinically diagnosed CRC and healthy individuals. Our results underline the potential association of the three major SCFA molecules (acetic, propionic, and butyric acid) with CRC risk and incidence.

Methods

We used Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 guideline [41, 42] to systematically search and extract data from primary human studies with SCFA measurement in CRC risk or incidence.
The Medline, Embase, and Web of Science database search was performed for articles involving human subjects that are in English from database conception until June 29, 2022. The details of the search keywords and strategies utilized in Ovid and Web of Science are available in the Additional file 1: Supplementary methods.

Eligibility criteria

All the records, including abstracts, were imported to EndNote X9 (Clarivate Analytics, Toronto, Canada). Duplicate records were first removed. The records were then filtered using EndNote’s built-in search tool for the following criteria: (i) searching for concentration*, level*, quanti*, measure*, assess*, evaluat*, estimat*, calculat*, mmol, and μmol as the inclusion criteria to capture studies which reported the SCFA measurement based on these terms, and (ii) searching for mouse, mice, murine, rats, conference, ethyl acetate (EtOAc), and phorbol as the exclusion criteria to exclude rodent studies, conference proceedings, and studies that have stated the use of any unrelated chemicals (such as EtOAc and 12-O-Tetradecanoylphorbol-13-acetate) - the asterisk symbol (*) applied was to include all the variations of the search terms. The abstracts of the remaining records were then screened to exclude reviews, methodology, human studies not related to SCFAs in CRC or CRA, and non-human studies (i.e., in vitro or other non-rodent animal studies), to identify the human studies on SCFA measurement in CRC or CRA. The full text of the remaining (n = 57) records were then screened to include only the observational studies which have measured fecal SCFA concentration. A final set of 23 observational studies qualified for further data extraction, quality assessment, and statistical analyses.

Data extraction and quality assessments

The data and additional details available for analysis (such as study subjects and SCFA levels) from the finalized primary studies were extracted and added to an Excel worksheet. The Newcastle-Ottawa Scale (NOS) [43] was used as a standard tool for quality assessment of 17 case-control studies in the selection, comparability, and exposure categories, to provide a score range between 0 and 9 (≤ 6, 7–8, and 9 indicate high, medium, and low risk of bias, respectively) [42]. Evaluation of six cross-sectional studies was performed using the Joanna Briggs Institute (JBI) Critical Appraisal Checklist tool [44], as recommended [45].

Statistical analyses

Review Manager (RevMan) software version 5.4 (Cochrane, Copenhagen, Denmark) was used to analyze the quantitative fecal SCFA concentration data, which were available in 10 of the final 23 observational studies (9 of 17 case-control, plus 1 of 6 cross-sectional studies). The fecal concentration of acetic, propionic, or butyric acid was considered as the subgroups. Before data entry, SEM or 95% CI upper and lower bound values were converted to SD. Due to variation in the reported SCFA concentration units between different papers, standardized mean difference (SMD) was selected as a measure of effect size for each study. The statistical heterogeneity among studies was calculated using χ2 and I2 tests and a P-value of 0.05 was considered significant [46]. To normalize the use of different SCFA measurement methods, a random-effects model was applied to analyze the pooled effect size and P-value for each SCFA molecule in each subgroup. One overall effect size and P-value of combined acetic, propionic, and butyric acid were also calculated. In all analyses, the effect size was reported with 95% confidence intervals, and the P-value < 0.05 was considered significant. Furthermore, the fixed-effect model was also applied in the case of non-significant heterogeneity of I2 < 50 [46]. All the data conversions, as well as qualitative and quantitative analyses, were validated by the second team member and confirmed by the senior authors.
One study [34] reported values that could not be converted to mean and SD for the meta-analysis and was therefore only included in our qualitative analysis. Another study [35] reported the numeric values of butyric acid concentration and other SCFA molecules in graphs and hence was included in both quantitative (for butyric acid) and qualitative data (for acetic acid, propionic acid and total SCFA). Therefore, in addition to studies in which the fecal SCFA concentration was presented using graphs (with no reported actual values), 14 of 23 studies were considered as qualitative studies (8 of 17 case-control, plus all 6 cross-sectional studies—including Ocvirk et al. 2020). The outcome of analyses from these qualitative studies was plotted as stacked bar charts, using Microsoft Excel (ver. 2016; Microsoft Corporation, Redmond, WA, USA).

Results

Study selection and quality assessment

The workflow on the identification and stepwise selection of the observational studies is presented in Fig. 1. Initially, a total of 2133 English language records obtained from searching through the three databases (Medline, Embase, and Web of Science) were imported to EndNote along with their abstracts. After removing duplicate records, the titles and abstracts of the remaining 1466 records were filtered and screened for eligibility as detailed in the Methods section. In total, 1409 records were excluded, of which most were in vitro studies. From the remaining 57 human studies, 34 studies were excluded. Of these 28 were interventional studies, three were observational studies on serum SCFA [4749], two studies had indistinct grouping (one case-control study with the presence of individuals with adenomatous polyps in the healthy control group [50] and one cross-sectional study with no clear definition of CRC high- and low-risk group [51]), and one retracted observational study [52].
Finally, 17 case-control and 6 cross-sectional studies were selected for data extraction and analysis. Table 1 summarizes the characteristics of these observational studies. The results of quality assessment using NOS and JBI tools on case-control and cross-sectional studies are provided in Additional file 1: Tables S1 and S2, respectively.
Table 1.
Characteristics of the selected studies. Cross-sectional studies are highlighted in gray, and case-control studies are not highlighted
https://static-content.springer.com/image/art%3A10.1186%2Fs12916-022-02529-4/MediaObjects/12916_2022_2529_Tab1_HTML.png
aCRC colorectal cancer, AP adenomatous polyposis, CD celiac disease, CRA colorectal adenoma, HC healthy controls, IBD inflammatory bowel disease. bGC-MS gas chromatography-mass spectrometry, HPLC high-performance liquid chromatography, FL fluorescence, 1H NMR 1H nuclear magnetic resonance spectroscopy, UPLC-MS ultra-performance liquid chromatography-tandem mass spectrometry, GLC gas-liquid chromatography. cC2 acetic acid, C3 propionic acid, C4 butyric acid. dRefer to the text for the definition of CRC risk and incidence category. eValues in this paper were measured on enema samples, not feces. Therefore, they used in qualitative analysis. fMore details are provided in the article. gRemoved from quantitative analysis as the reported SCFA values could not be converted to mean and SD. hRemoved from meta-analysis due to insufficient data on SCFA measurement method. iSCFAs were measured in only a subset of these subjects (n = 25 large/small adenoma and n = 23 adenoma-free). jCombined values of males and females

Stratifications based on CRC risk or incidence

Studies listed in Table 1 are presented based on the type of data provided (qualitative or quantitative) and CRC risk and/or incidence. Among the 17 case-control studies (not highlighted in Table 1), 8 studies comparing CRC cases and healthy control subjects were allocated to the CRC incidence category, 5 studies comparing individuals with CRA and healthy controls assigned to the CRC risk category, and the remaining 4 studies were included in both incidence and risk categories since they compared CRC patients, CRA individuals, and healthy subjects. All 6 cross-sectional studies (highlighted gray in Table 1) comparing populations with high- versus low-risk of CRC were allocated to the risk category. Therefore, the CRC incidence and risk category included 12 and 15 studies, respectively (Table 1). For each study, the details of the measured SCFA and CRC risk and/or incidence grouping are provided in Additional file 1: Table S3. Some studies reported total SCFA concentration in addition to the individual (acetate, propionate, and butyrate) SCFAs.
The primary studies analyzed in this systematic review were performed in various countries and ethnic groups. Age was matched in some of the studies [18, 24, 28, 3840], although the male-to-female ratio was not similar between the study groups in most studies (Table 1). The SCFA concentrations were measured using different techniques, such as gas chromatography, liquid chromatography, gas-liquid chromatography and 1H nuclear magnetic resonance spectroscopy.

Data analyses

The meta-analysis of the quantitative data extracted from the 10 selected studies [1822, 26, 28, 30, 33, 35] are presented in Fig. 2. In the risk category (Fig. 2A. B), two studies [19, 20] were excluded from the meta-analysis due to the lack of sufficient details of the methods used for SCFA measurement from stool samples. In CRC risk meta-analysis, the effect size of each of the three SCFAs was not statistically significant; however, their combined effect size was significantly higher in low risk compared to high-risk CRC (SMD = 2.02, 95% CI 0.31 to 3.74, P = 0.02, Fig. 2A). The effect size of total SCFA concentration was not statistically significant in the low- vs high-risk group (Fig. 2B).
In the CRC incidence analysis (Fig. 2C), the fecal concentrations of acetic acid (SMD = 0.61, 95% CI 0.09 to 1.13, P = 0.02) and butyric acid (SMD = 0.45, 95% CI 0.02 to 0.88, P = 0.04) were significantly higher in the healthy control compared to CRC cases. In addition, the combined effect size of acetic, propionic, and butyric acid remained significant between CRC cases and healthy controls (SMD = 0.45, 95% CI 0.19 to 0.72, P = 0.0009, Fig. 2C).
Furthermore, the I2 heterogeneity index was in the “moderate” range (30 to 60%) [46] for the meta-analysis of total SCFA concentration in CRC risk (Fig. 2B) and butyric acid in CRC incidence (Fig. 2C) category. Therefore, we performed another meta-analysis using the fixed-effect model on the same data instead of the random-effect model presented in Table 2. This resulted in a more pronounced difference in butyric acid concentration between CRC cases and healthy controls (SMD = 0.42, 95% CI 0.1 to 0.74, P = 0.009). The results of the fixed-effect model meta-analyses are presented in Additional file 1: Fig. S1 and S2, respectively, and the findings of all quantitative meta-analyses are summarized in Table 2.
Table 2
Summary of the outcomes of each meta-analysis. Significant P values of the effect size are in bold
 
Measured SCFA
Number of Studies
Heterogeneity
(I2 %, P value)
Statistical Model
Effect size (SMD [95% CI], P value)
CRC risk
Acetic acid
3
99, < 0.00001
Random effect
2.63 [− 1.82 to 7.08], 0.25
Propionic acid
3
99, < 0.00001
Random effect
2.33 [− 2.33 to 7.00], 0.33
Butyric acid
5
99, < 0.00001
Random effect
1.47 [− 0.80 to 3.73], 0.2
Combineda
11
99, < 0.00001
Random effect
2.02 [0.31 to 3.74], 0.02
Total SCFA
3
38, 0.2
Random effect
0.29 [− 0.13 to 0.72], 0.17
Total SCFA
3
38, 0.2
Fixed effect
0.25 [− 0.05 to 0.56], 0.11
CRC incidence
Acetic acid
4
60, 0.06
Random effect
0.61 [0.09 to 1.13], 0.02
Propionic acid
4
62, 0.05
Random effect
0.32 [− 0.21 to 0.84], 0.24
Butyric acid
4
43, 0.15
Random effect
0.45 [0.02 to 0.88], 0.04
Combineda
12
51, 0.02
Random effect
0.45 [0.19 to 0.72], 0.0009
Butyric acid
4
43, 0.15
Fixed effect
0.42 [0.1 to 0.74], 0.009
aCombined effect size of acetic, propionic, and butyric acid. Note that the total SCFA indicates the collection of all the SCFA molecules—not only acetic, propionic, and butyric acid
Qualitative analysis was carried out on the studies which reported lower, higher or no changes to the concentration of SCFAs between high-risk CRC (for risk category) or CRC case (for incidence) and low-risk or control, respectively [2325, 27, 29, 31, 32, 3440] (Additional file 1: Fig. S3). In the risk category, more studies (70.4%) reported significantly lower concentrations of fecal acetic, propionic, and butyric acid as well as total SCFA in individuals at high risk of CRC. In the incidence category, more studies (66.7%) reported significantly lower concentrations of fecal acetic and butyric acid in CRC patients compared to healthy controls. However, the number of studies reporting no significant difference in the propionic acid was the highest in the incidence category. Overall, our qualitative analysis (Additional file 1: Fig. S3) corroborates with the meta-analysis results (Fig. 2).

Discussion

For more than three decades, in vitro, animal, and human studies have identified numerous potentially beneficial anti-inflammatory and anti-carcinogenic roles of SCFA molecules in gut health and colonic diseases [6, 1417, 53]. In addition, several meta-analyses (Additional file 1: Table S4) have assessed the role of colonic microbiota [54], non-digestible carbohydrates [55] and dietary fiber in colorectal carcinoma [11, 56] or adenoma [9, 10] as well as the alteration of SCFAs in irritable bowel syndrome (IBS) [57], or inflammatory bowel disease (IBD) [58].
This systematic review and meta-analysis were conducted on 23 studies to better determine the potential association between fecal SCFA concentration and CRC risk and incidence. The combined mean difference of acetic, propionic, and butyric acid in the CRC risk category analysis revealed a significantly lower concentration of these SCFAs in individuals at risk of developing CRC compared to healthy subjects, indicating a potential association between these three major SCFA molecules and CRC development. This finding was further confirmed in the CRC incidence category analysis where the fecal SCFA levels in CRC patients were significantly lower compared to those in healthy subjects.
Our findings in CRC risk and incidence were consistent with the observations reported in other meta-analyses, which focused on the association between dietary fiber intake and the risk of colorectal adenoma [9, 10], and carcinoma [11]. These systematic reviews suggested a protective effect of dietary fiber intake against CRA and CRC [911]. Since SCFAs are produced by gut-microbiota via the fermentation of dietary fibers [1417], our meta-analysis of SCFA concentrations in CRC further confirms earlier observations and underlines the importance of dietary fibers/SCFAs in the risk and progression of CRC.
Another meta-analysis, which assessed the effect of non-digestible carbohydrate [resistance starch (RS)] or inulin supplementation on the risk of colorectal neoplasia, did not find significant increase in fecal total SCFA or butyric acid concentration and excretion before and after the intervention [55]. Many studies which investigated the effect of RS on healthy subjects or individuals with sporadic CRC or adenoma had a period of ≤ 4-week of intervention. A few studies reported 7- and 8-week intervention on adenoma or healthy individuals and the remaining studies were conducted on individuals with inherited CRC syndromes after > 2-year intervention [55]. The duration of intervention was longest (> 2 years) for studies involving hereditary CRC cases with reported germ-line mutations, which may have outweighed the effect of RS supplementation, while interventions involving sporadic cases or healthy subjects had much shorter periods of RS intervention (< 8 weeks) [55]. In our meta-analysis, we also did not observe a significant difference in total fecal SCFAs in the CRC risk category. This could be due to other SCFA molecules such as valeric, iso-butyric, and iso-valeric acid being included in total SCFA measurements; the latter two are the branched SCFAs mainly produced via fermentation of branched amino acids in the colon and not from non-digestible carbohydrates [59, 60].
Another systematic review on the food-microorganism-SCFA axis, without any meta-analysis, concluded that most evidence demonstrated higher SCFA levels in individuals at risk of CRC compared to healthy individuals [61], which contrasts with findings in our systematic review which showed lower fecal SCFA concentration in at-risk individuals (Fig. 2). In comparison to our systematic review, their search strategy restricted their analysis to only 8 of the final 23 studies that we analyzed [19, 27, 31, 32, 3740]. Therefore, their conclusion was based on a smaller subset of the primary studies available and was also not supported by a meta-analysis.
Both the quantitative and qualitative analyses of CRC risk identified comparable findings of significantly lower concentration of acetic and butyric acid in the high- versus low-risk CRC group. For the CRC incidence category, the quantitative meta-analysis of butyric acid was consistent with observations identified in most of the articles from the qualitative analysis, supporting the evidence of lower concentration of three SCFAs in CRC cases compared to healthy controls. The meta-analysis of propionic acid was not significantly different between cases and controls. Similarly, most of the studies (5 of 7) reported no significant difference in fecal propionic acid concentration between CRC and healthy control in the qualitative analysis. The meta-analysis on IBS revealed a significantly higher concentration of fecal propionic acid in these patients in comparison to healthy controls [57]. Therefore, further studies comparing SCFA profiles among multiple gut diseases could shed more light on the importance of these molecules in the development of varied medical conditions.
To our knowledge, this systematic review is the first to provide a comprehensive search and data collection on observational studies linking SCFA molecules with the CRC risk and incidence. A limitation of our analysis is the heterogeneity of the studies evaluated in this systematic review, which is very difficult to control for. One such factor was the age group assessed for CRC incidence and risk. The mean age of the group in the studies was greater than 50 years and fecal SCFA concentration was not measured in younger populations to provide a comparison with low-risk, young age individuals. Although CRC is most often diagnosed in individuals > 50 years, the incidence for early-onset CRC (EOCRC) in adults aged 20–49 years has increased over the past decade in the USA, Australia, and Europe [6265]. It would be of interest in the future to study different age group populations for CRC risk and incidence. Family history [66], diet, and lifestyle [67] are known factors contributing to CRC incidence. Only a few studies assessed in this systematic review provided information on the dietary difference between groups [19, 21, 35, 36, 40]. There was also no information about the type of polyps (conventional vs serrated) in individuals with CRA.
One common limitation is related to the nature of observational studies. Case-control studies are inherently prone to recall bias and appropriate matching of case and control groups [68]. Similarly, the results of the cross-sectional studies could be affected by bidirectional relationship [69] and confounding factors. We have assessed the effect of these inherent limitations on our analysis by undertaking appropriate quality checks such as the “comparability” category of NOS quality assessment (Additional file 1: Table S1) and the JBI tool (Additional file 1: Table S2) for all the studies included in our systematic review and meta-analysis.
Another limitation is the diversity in sample handling/storage workflows and the methodologies used to measure fecal SCFA across the studies (Table 1). We did not have enough studies to perform separate meta-analyses to understand the effect of each of these variables on the SCFA concentrations. While these factors could influence our interpretation, the levels of SCFAs were lower in high-risk as well as incident CRC cases, irrespective of the method used to measure SCFAs. Since these are well-known and established techniques for measuring SCFA concentrations, it appears that the standardized sample handling workflows and analytical methods had little impact on differences across study groups, so long as optimized procedures for SCFA assessment were followed. This systematic review did not include non-English records. To our knowledge, no longitudinal studies have reported fecal SCFA measurements at different time points during CRC progression, nonetheless, the 23 studies assessed in this systematic review and meta-analysis provide a comparison between CRC risk/incidence and respective controls from various countries and ethnic groups.
In addition to the SCFAs assessed in this systematic review, other metabolites such as bile acids were also measured in six of the selected studies [20, 30, 32, 35, 36, 39]. Among the bile acids investigated, a significantly higher fecal concentration of deoxycholic acid in the CRC high- versus low-risk group was reported in three studies [35, 36, 39]. Dietary fiber and fat promote the production of SCFA and bile acid molecules in the gut, respectively, and the latter is associated with gastrointestinal carcinogenesis [7072]. Measurement of fecal SCFAs and other gut metabolites (such as bile acids) in longitudinal studies comparing individuals with colorectal adenoma/risk and healthy subjects could strengthen their association with CRC progression.
This study supports further exploration into fecal concentration of SCFAs: acetic, propionic, and butyric acids, as biomarkers for CRC risk. Among the current CRC screening methods, colonoscopy is the gold standard [73]; however, being invasive, it presents some procedural risk [74]. The guaiac fecal occult blood test (gFOBT) and fecal immunochemical test (FIT) are other, in practice, non-invasive stool-based methods for CRC screening, which however require improvement, in particular for detection of CRA or early-stage colonic carcinogenesis [7476]. Fecal SCFA could be considered as a potential non-invasive biomarker to be measured in combination with or as an alternative to the commonly used non-invasive and current CRC screening methods [74, 77], to improve specificity and sensitivity of current screening, as well as for potential early detection of CRA.

Conclusions

Gut microbiota dysbiosis and changes in their metabolites have been the focus of epidemiological studies aimed at uncovering associations with colonic inflammation and carcinogenesis. In line with the protective role of fecal SCFAs against the development of gut diseases [15, 16], and the protective effect of dietary fibers against CRC risk and/or incidence [911], we determined that the combined fecal concentration of the three major SCFA molecules was significantly lower not only in CRC patients compared to healthy controls, but also in high-risk CRC individuals. Gut SCFA concentrations are inversely associated with CRC-risk as well as CRC-incidence and could be biomarkers for predicting CRC-progression, as well as a drug target (in future intervention studies) aimed to retard or prevent CRC progression.

Registration

The study is registered in PROSPERO database (registration code: CRD42021256123).

Acknowledgements

The authors acknowledge Dr. Ho Pham for his assistance and the infrastructure support through the School of Medicine, Western Sydney University. The authors also acknowledge Mrs. Ameneh Najdi for cross-checking the data entry in the analyses.

Declarations

Not applicable.
Not applicable; this manuscript does not include any details, images, or videos relating to an individual person.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.PubMedCrossRef Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.PubMedCrossRef
2.
Zurück zum Zitat Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global patterns and trends in colorectal cancer incidence and mortality. Gut. 2017;66(4):683–91.PubMedCrossRef Arnold M, Sierra MS, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global patterns and trends in colorectal cancer incidence and mortality. Gut. 2017;66(4):683–91.PubMedCrossRef
3.
Zurück zum Zitat Nguyen LH, Goel A, Chung DC. Pathways of colorectal carcinogenesis. Gastroenterology. 2020;158(2):291–302.PubMedCrossRef Nguyen LH, Goel A, Chung DC. Pathways of colorectal carcinogenesis. Gastroenterology. 2020;158(2):291–302.PubMedCrossRef
4.
Zurück zum Zitat Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB. Colorectal cancer. Lancet. 2019;394(10207):1467–80.PubMedCrossRef Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB. Colorectal cancer. Lancet. 2019;394(10207):1467–80.PubMedCrossRef
5.
Zurück zum Zitat Mármol I, Sánchez-De-Diego C, Pradilla Dieste A, Cerrada E, Rodriguez YM. Colorectal carcinoma: a general overview and future perspectives in colorectal cancer. Int J Mol Sci. 2017;18(1):197.PubMedCentralCrossRef Mármol I, Sánchez-De-Diego C, Pradilla Dieste A, Cerrada E, Rodriguez YM. Colorectal carcinoma: a general overview and future perspectives in colorectal cancer. Int J Mol Sci. 2017;18(1):197.PubMedCentralCrossRef
6.
Zurück zum Zitat Wong SH, Yu J. Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol. 2019;16(11):690–704.PubMedCrossRef Wong SH, Yu J. Gut microbiota in colorectal cancer: mechanisms of action and clinical applications. Nat Rev Gastroenterol Hepatol. 2019;16(11):690–704.PubMedCrossRef
7.
Zurück zum Zitat Zhao Z, Feng Q, Yin Z, Shuang J, Bai B, Yu P, et al. Red and processed meat consumption and colorectal cancer risk: a systematic review and meta-analysis. Oncotarget. 2017;8(47):83306–14.PubMedPubMedCentralCrossRef Zhao Z, Feng Q, Yin Z, Shuang J, Bai B, Yu P, et al. Red and processed meat consumption and colorectal cancer risk: a systematic review and meta-analysis. Oncotarget. 2017;8(47):83306–14.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Chan DS, Lau R, Aune D, Vieira R, Greenwood DC, Kampman E, et al. Red and processed meat and colorectal cancer incidence: meta-analysis of prospective studies. PLoS One. 2011;6(6):e20456.PubMedPubMedCentralCrossRef Chan DS, Lau R, Aune D, Vieira R, Greenwood DC, Kampman E, et al. Red and processed meat and colorectal cancer incidence: meta-analysis of prospective studies. PLoS One. 2011;6(6):e20456.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Nucci D, Fatigoni C, Salvatori T, Nardi M, Realdon S, Gianfredi V. Association between dietary fibre intake and colorectal adenoma: a systematic review and meta-analysis. Int J Environ Res Public Health. 2021;18(8):4168. Nucci D, Fatigoni C, Salvatori T, Nardi M, Realdon S, Gianfredi V. Association between dietary fibre intake and colorectal adenoma: a systematic review and meta-analysis. Int J Environ Res Public Health. 2021;18(8):4168.
10.
Zurück zum Zitat Oh H, Kim H, Lee DH, Lee A, Giovannucci EL, Kang SS, et al. Different dietary fibre sources and risks of colorectal cancer and adenoma: a dose-response meta-analysis of prospective studies. Br J Nutr. 2019;122(6):605–15.PubMedCrossRef Oh H, Kim H, Lee DH, Lee A, Giovannucci EL, Kang SS, et al. Different dietary fibre sources and risks of colorectal cancer and adenoma: a dose-response meta-analysis of prospective studies. Br J Nutr. 2019;122(6):605–15.PubMedCrossRef
11.
Zurück zum Zitat Gianfredi V, Salvatori T, Villarini M, Moretti M, Nucci D, Realdon S. Is dietary fibre truly protective against colon cancer? A systematic review and meta-analysis. Int J Food Sci Nutr. 2018;69(8):904–15.PubMedCrossRef Gianfredi V, Salvatori T, Villarini M, Moretti M, Nucci D, Realdon S. Is dietary fibre truly protective against colon cancer? A systematic review and meta-analysis. Int J Food Sci Nutr. 2018;69(8):904–15.PubMedCrossRef
13.
Zurück zum Zitat 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
14.
Zurück zum Zitat Alexander C, Swanson KS, Fahey GC Jr, Garleb KA. Perspective: Physiologic importance of short-chain fatty acids from nondigestible carbohydrate fermentation. Adv Nutr. 2019;10(4):576–89.PubMedPubMedCentralCrossRef Alexander C, Swanson KS, Fahey GC Jr, Garleb KA. Perspective: Physiologic importance of short-chain fatty acids from nondigestible carbohydrate fermentation. Adv Nutr. 2019;10(4):576–89.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Parada Venegas D, De la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, et al. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 2019;10(277):1-16. Parada Venegas D, De la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, et al. Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 2019;10(277):1-16.
16.
Zurück zum Zitat Liu P, Wang Y, Yang G, Zhang Q, Meng L, Xin Y, et al. The role of short-chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and colonic carcinogenesis. Pharmacol Res. 2021;165:105420.PubMedCrossRef Liu P, Wang Y, Yang G, Zhang Q, Meng L, Xin Y, et al. The role of short-chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and colonic carcinogenesis. Pharmacol Res. 2021;165:105420.PubMedCrossRef
17.
Zurück zum Zitat van der Beek CM, Dejong CHC, Troost FJ, Masclee AAM, Lenaerts K. Role of short-chain fatty acids in colonic inflammation, carcinogenesis, and mucosal protection and healing. Nutr Rev. 2017;75(4):286–305.PubMedCrossRef van der Beek CM, Dejong CHC, Troost FJ, Masclee AAM, Lenaerts K. Role of short-chain fatty acids in colonic inflammation, carcinogenesis, and mucosal protection and healing. Nutr Rev. 2017;75(4):286–305.PubMedCrossRef
18.
Zurück zum Zitat Boutron-Ruault MC, Marteau P, Lavergne-Slove A, Myara A, Gerhardt MF, Franchisseur C, et al. Effects of a 3-mo consumption of short-chain fructo-oligosaccharides on parameters of colorectal carcinogenesis in patients with or without small or large colorectal adenomas. Nutr Cancer. 2005;53(2):160–8.PubMedCrossRef Boutron-Ruault MC, Marteau P, Lavergne-Slove A, Myara A, Gerhardt MF, Franchisseur C, et al. Effects of a 3-mo consumption of short-chain fructo-oligosaccharides on parameters of colorectal carcinogenesis in patients with or without small or large colorectal adenomas. Nutr Cancer. 2005;53(2):160–8.PubMedCrossRef
19.
Zurück zum Zitat Bridges KM, Diaz FJ, Wang Z, Ahmed I, Sullivan DK, Umar S, et al. Relating stool microbial metabolite levels, inflammatory markers and dietary behaviors to screening colonoscopy findings in a racially/ethnically diverse patient population. Genes. 2018;9(3):119. Bridges KM, Diaz FJ, Wang Z, Ahmed I, Sullivan DK, Umar S, et al. Relating stool microbial metabolite levels, inflammatory markers and dietary behaviors to screening colonoscopy findings in a racially/ethnically diverse patient population. Genes. 2018;9(3):119.
20.
Zurück zum Zitat Chen CY, Niu M, Pan JX, Du N, Liu SM, Li HQ, et al. Bacteroides, butyric acid and t10,c12-CLA changes in colorectal adenomatous polyp patients. Gut Pathogens. 2021;13(1):9.CrossRef Chen CY, Niu M, Pan JX, Du N, Liu SM, Li HQ, et al. Bacteroides, butyric acid and t10,c12-CLA changes in colorectal adenomatous polyp patients. Gut Pathogens. 2021;13(1):9.CrossRef
21.
Zurück zum Zitat Chen HM, Yu YN, Wang JL, Lin YW, Kong X, Yang CQ, et al. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am J Clin Nutr. 2013;97(5):1044–52.PubMedCrossRef Chen HM, Yu YN, Wang JL, Lin YW, Kong X, Yang CQ, et al. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am J Clin Nutr. 2013;97(5):1044–52.PubMedCrossRef
22.
Zurück zum Zitat Kashtan H, Stern HS, Jenkins DJA, Jenkins AL, Thompson LU, Hay K, et al. Colonic fermentation and markers of colorectal-cancer risk. Am J Clin Nutr. 1992;55(3):723–8.PubMedCrossRef Kashtan H, Stern HS, Jenkins DJA, Jenkins AL, Thompson LU, Hay K, et al. Colonic fermentation and markers of colorectal-cancer risk. Am J Clin Nutr. 1992;55(3):723–8.PubMedCrossRef
23.
Zurück zum Zitat Lin Y, Ma C, Liu C, Wang Z, Yang J, Liu X, et al. NMR-based fecal metabolomics fingerprinting as predictors of earlier diagnosis in patients with colorectal cancer. Oncotarget. 2016;7(20):29454–64.PubMedPubMedCentralCrossRef Lin Y, Ma C, Liu C, Wang Z, Yang J, Liu X, et al. NMR-based fecal metabolomics fingerprinting as predictors of earlier diagnosis in patients with colorectal cancer. Oncotarget. 2016;7(20):29454–64.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Lin Y, Ma CC, Bezabeh T, Wang ZN, Liang JH, Huang Y, et al. H-1 NMR-based metabolomics reveal overlapping discriminatory metabolites and metabolic pathway disturbances between colorectal tumor tissues and fecal samples. Int J Cancer. 2019;145(6):1679–89.PubMedCrossRef Lin Y, Ma CC, Bezabeh T, Wang ZN, Liang JH, Huang Y, et al. H-1 NMR-based metabolomics reveal overlapping discriminatory metabolites and metabolic pathway disturbances between colorectal tumor tissues and fecal samples. Int J Cancer. 2019;145(6):1679–89.PubMedCrossRef
25.
Zurück zum Zitat Monleon D, Morales JM, Barrasa A, Lopez JA, Vazquez C, Celda B. Metabolite profiling of fecal water extracts from human colorectal cancer. NMR Biomed. 2009;22(3):342–8.PubMedCrossRef Monleon D, Morales JM, Barrasa A, Lopez JA, Vazquez C, Celda B. Metabolite profiling of fecal water extracts from human colorectal cancer. NMR Biomed. 2009;22(3):342–8.PubMedCrossRef
26.
Zurück zum Zitat Niccolai E, Baldi S, Ricci F, Russo E, Nannini G, Menicatti M, et al. Evaluation and comparison of short chain fatty acids composition in gut diseases. World J Gastroenterol. 2019;25(36):5543–58.PubMedPubMedCentralCrossRef Niccolai E, Baldi S, Ricci F, Russo E, Nannini G, Menicatti M, et al. Evaluation and comparison of short chain fatty acids composition in gut diseases. World J Gastroenterol. 2019;25(36):5543–58.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Ohigashi S, Sudo K, Kobayashi D, Takahashi O, Takahashi T, Asahara T, et al. Changes of the intestinal microbiota, short chain fatty acids, and fecal pH in patients with colorectal cancer. Dig Dis Sci. 2013;58(6):1717–26.PubMedCrossRef Ohigashi S, Sudo K, Kobayashi D, Takahashi O, Takahashi T, Asahara T, et al. Changes of the intestinal microbiota, short chain fatty acids, and fecal pH in patients with colorectal cancer. Dig Dis Sci. 2013;58(6):1717–26.PubMedCrossRef
28.
Zurück zum Zitat Song EM, Byeon JS, Lee SM, Yoo HJ, Kim SJ, Lee SH, et al. Fecal fatty acid profiling as a potential new screening biomarker in patients with colorectal cancer. Dig Dis Sci. 2018;63(5):1229–36.PubMedCrossRef Song EM, Byeon JS, Lee SM, Yoo HJ, Kim SJ, Lee SH, et al. Fecal fatty acid profiling as a potential new screening biomarker in patients with colorectal cancer. Dig Dis Sci. 2018;63(5):1229–36.PubMedCrossRef
29.
Zurück zum Zitat Sze MA, Topcuoglu BD, Lesniak NA, Ruffin MT, Schloss PD. Fecal short-chain fatty acids are not predictive of colonic tumor status and cannot be predicted based on bacterial community structure. mBio. 2019;10(4):e01454-19. Sze MA, Topcuoglu BD, Lesniak NA, Ruffin MT, Schloss PD. Fecal short-chain fatty acids are not predictive of colonic tumor status and cannot be predicted based on bacterial community structure. mBio. 2019;10(4):e01454-19.
30.
Zurück zum Zitat Torii T, Kanemitsu K, Hagiwara A. Simultaneous assay of fecal short-chain fatty and bile acids and ratio of total bile acids to butyrate in colon cancer. Chromatography. 2019;40(2):49–57.CrossRef Torii T, Kanemitsu K, Hagiwara A. Simultaneous assay of fecal short-chain fatty and bile acids and ratio of total bile acids to butyrate in colon cancer. Chromatography. 2019;40(2):49–57.CrossRef
31.
Zurück zum Zitat Weaver GA, Krause JA, Miller TL, Wolin MJ. Short chain fatty acid distributions of enema samples from a sigmoidoscopy population: an association of high acetate and low butyrate ratios with adenomatous polyps and colon cancer. Gut. 1988;29(11):1539–43.PubMedPubMedCentralCrossRef Weaver GA, Krause JA, Miller TL, Wolin MJ. Short chain fatty acid distributions of enema samples from a sigmoidoscopy population: an association of high acetate and low butyrate ratios with adenomatous polyps and colon cancer. Gut. 1988;29(11):1539–43.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Weir TL, Manter DK, Sheflin AM, Barnett BA, Heuberger AL, Ryan EP. Stool microbiome and metabolome differences between colorectal cancer patients and healthy adults. PLoS One. 2013;8(8):e70803. Weir TL, Manter DK, Sheflin AM, Barnett BA, Heuberger AL, Ryan EP. Stool microbiome and metabolome differences between colorectal cancer patients and healthy adults. PLoS One. 2013;8(8):e70803.
33.
Zurück zum Zitat Yusuf F, Adewiah S, Fatchiyah F. The level short chain fatty acids and HSP 70 in colorectal cancer and non-colorectal cancer. Acta Informatica Medica. 2018;26(3):160–3.PubMedPubMedCentralCrossRef Yusuf F, Adewiah S, Fatchiyah F. The level short chain fatty acids and HSP 70 in colorectal cancer and non-colorectal cancer. Acta Informatica Medica. 2018;26(3):160–3.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Nannini G, Meoni G, Tenori L, Ringressi MN, Taddei A, Niccolai E, et al. Fecal metabolomic profiles: a comparative study of patients with colorectal cancer vs adenomatous polyps. World J Gastroenterol. 2021;27(38):6430–41.PubMedPubMedCentralCrossRef Nannini G, Meoni G, Tenori L, Ringressi MN, Taddei A, Niccolai E, et al. Fecal metabolomic profiles: a comparative study of patients with colorectal cancer vs adenomatous polyps. World J Gastroenterol. 2021;27(38):6430–41.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Ocvirk S, Posma JM, Li JV, Koller KR, Day GM, Flanagan CA, et al. A prospective cohort analysis of gut microbial co-metabolism in Alaska Native and rural African people at high and low risk of colorectal cancer. Am J Clin Nutr. 2020;111(2):406–19.PubMedCrossRef Ocvirk S, Posma JM, Li JV, Koller KR, Day GM, Flanagan CA, et al. A prospective cohort analysis of gut microbial co-metabolism in Alaska Native and rural African people at high and low risk of colorectal cancer. Am J Clin Nutr. 2020;111(2):406–19.PubMedCrossRef
36.
Zurück zum Zitat Katsidzira L, Ocvirk S, Wilson A, Li J, Mahachi CB, Soni D, et al. Differences in fecal gut microbiota, short-chain fatty acids and bile acids link colorectal cancer risk to dietary changes associated with urbanization among Zimbabweans. Nutr Cancer. 2019;71(8):1313–24.PubMedCrossRef Katsidzira L, Ocvirk S, Wilson A, Li J, Mahachi CB, Soni D, et al. Differences in fecal gut microbiota, short-chain fatty acids and bile acids link colorectal cancer risk to dietary changes associated with urbanization among Zimbabweans. Nutr Cancer. 2019;71(8):1313–24.PubMedCrossRef
37.
Zurück zum Zitat Hester CM, Jala VR, Langille MGI, Umar S, Greiner KA, Haribabu B. Fecal microbes, short chain fatty acids, and colorectal cancer across racial/ethnic groups. World J Gastroenterol. 2015;21(9):2759–69.PubMedPubMedCentralCrossRef Hester CM, Jala VR, Langille MGI, Umar S, Greiner KA, Haribabu B. Fecal microbes, short chain fatty acids, and colorectal cancer across racial/ethnic groups. World J Gastroenterol. 2015;21(9):2759–69.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Ou J, Carbonero F, Zoetendal EG, DeLany JP, Wang M, Newton K, et al. Diet, microbiota, and microbial metabolites in colon cancer risk in rural Africans and African Americans. Am J Clin Nutr. 2013;98(1):111–20.PubMedPubMedCentralCrossRef Ou J, Carbonero F, Zoetendal EG, DeLany JP, Wang M, Newton K, et al. Diet, microbiota, and microbial metabolites in colon cancer risk in rural Africans and African Americans. Am J Clin Nutr. 2013;98(1):111–20.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Ou J, DeLany JP, Zhang M, Sharma S, O'Keefe SJD. Association between low colonic short-chain fatty acids and high bile acids in high colon cancer risk populations. Nutr Cancer. 2012;64(1):34–40.PubMedCrossRef Ou J, DeLany JP, Zhang M, Sharma S, O'Keefe SJD. Association between low colonic short-chain fatty acids and high bile acids in high colon cancer risk populations. Nutr Cancer. 2012;64(1):34–40.PubMedCrossRef
40.
Zurück zum Zitat O'Keefe SJD, Ou J, Aufreiter S, O'Connor D, Sharma S, Sepulveda J, et al. Products of the colonic microbiota mediate the effects of diet on colon cancer risk. J Nutr. 2009;139(11):2044–8.PubMedPubMedCentralCrossRef O'Keefe SJD, Ou J, Aufreiter S, O'Connor D, Sharma S, Sepulveda J, et al. Products of the colonic microbiota mediate the effects of diet on colon cancer risk. J Nutr. 2009;139(11):2044–8.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. Bmj. 2021;372:n71.PubMedPubMedCentralCrossRef Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. Bmj. 2021;372:n71.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Muka T, Glisic M, Milic J, Verhoog S, Bohlius J, Bramer W, et al. A 24-step guide on how to design, conduct, and successfully publish a systematic review and meta-analysis in medical research. Eur J Epidemiol. 2020;35(1):49–60.PubMedCrossRef Muka T, Glisic M, Milic J, Verhoog S, Bohlius J, Bramer W, et al. A 24-step guide on how to design, conduct, and successfully publish a systematic review and meta-analysis in medical research. Eur J Epidemiol. 2020;35(1):49–60.PubMedCrossRef
44.
45.
Zurück zum Zitat Ma LL, Wang YY, Yang ZH, Huang D, Weng H, Zeng XT. Methodological quality (risk of bias) assessment tools for primary and secondary medical studies: what are they and which is better? Mil Med Res. 2020;7(1):7.PubMedPubMedCentral Ma LL, Wang YY, Yang ZH, Huang D, Weng H, Zeng XT. Methodological quality (risk of bias) assessment tools for primary and secondary medical studies: what are they and which is better? Mil Med Res. 2020;7(1):7.PubMedPubMedCentral
47.
Zurück zum Zitat Baldi S, Menicatti M, Nannini G, Niccolai E, Russo E, Ricci F, et al. Free fatty acids signature in human intestinal disorders: Significant association between butyric acid and celiac disease. Nutrients. 2021;13(3):1–14.CrossRef Baldi S, Menicatti M, Nannini G, Niccolai E, Russo E, Ricci F, et al. Free fatty acids signature in human intestinal disorders: Significant association between butyric acid and celiac disease. Nutrients. 2021;13(3):1–14.CrossRef
48.
Zurück zum Zitat Bartolucci G, Pallecchi M, Menicatti M, Moracci L, Pucciarelli S, Agostini M, et al. A method for assessing plasma free fatty acids from C2 to C18 and its application for the early detection of colorectal cancer. J Pharm Biomed Anal. 2022;215:114762.PubMedCrossRef Bartolucci G, Pallecchi M, Menicatti M, Moracci L, Pucciarelli S, Agostini M, et al. A method for assessing plasma free fatty acids from C2 to C18 and its application for the early detection of colorectal cancer. J Pharm Biomed Anal. 2022;215:114762.PubMedCrossRef
49.
Zurück zum Zitat Genua F, Mirković B, Mullee A, Levy M, Gallagher WM, Vodicka P, et al. Association of circulating short chain fatty acid levels with colorectal adenomas and colorectal cancer. Clin Nutr ESPEN. 2021;46:297–304.PubMedCrossRef Genua F, Mirković B, Mullee A, Levy M, Gallagher WM, Vodicka P, et al. Association of circulating short chain fatty acid levels with colorectal adenomas and colorectal cancer. Clin Nutr ESPEN. 2021;46:297–304.PubMedCrossRef
50.
Zurück zum Zitat Amiot A, Dona AC, Wijeyesekera A, Tournigand C, Baumgaertner I, Lebaleur Y, et al. H-1 NMR spectroscopy of fecal extracts enables detection of advanced colorectal neoplasia. J Proteome Res. 2015;14(9):3871–81.PubMedCrossRef Amiot A, Dona AC, Wijeyesekera A, Tournigand C, Baumgaertner I, Lebaleur Y, et al. H-1 NMR spectroscopy of fecal extracts enables detection of advanced colorectal neoplasia. J Proteome Res. 2015;14(9):3871–81.PubMedCrossRef
51.
Zurück zum Zitat Segal I, Hassan H, Walker ARP, Becker P, Braganza J. Fecal short chain fatty acids in South African urban Africans and whites. Dis Colon Rectum. 1995;38(7):732–4.PubMedCrossRef Segal I, Hassan H, Walker ARP, Becker P, Braganza J. Fecal short chain fatty acids in South African urban Africans and whites. Dis Colon Rectum. 1995;38(7):732–4.PubMedCrossRef
52.
Zurück zum Zitat Wang X, Wang J, Rao B, Deng LI. Gut flora profiling and fecal metabolite composition of colorectal cancer patients and healthy individuals. Exper Therapeutic Med. 2017;13(6):2848–54.CrossRef Wang X, Wang J, Rao B, Deng LI. Gut flora profiling and fecal metabolite composition of colorectal cancer patients and healthy individuals. Exper Therapeutic Med. 2017;13(6):2848–54.CrossRef
53.
Zurück zum Zitat Song M, Chan AT, Sun J. Influence of the gut microbiome, diet, and environment on risk of colorectal cancer. Gastroenterology. 2020;158(2):322–40.PubMedCrossRef Song M, Chan AT, Sun J. Influence of the gut microbiome, diet, and environment on risk of colorectal cancer. Gastroenterology. 2020;158(2):322–40.PubMedCrossRef
54.
Zurück zum Zitat Borges-Canha M, Portela-Cidade JP, Dinis-Ribeiro M, Leite-Moreira AF, Pimentel-Nunes P. Role of colonic microbiota in colorectal carcinogenesis: a systematic review. Rev Esp Enferm Dig. 2015;107(11):659–71.PubMed Borges-Canha M, Portela-Cidade JP, Dinis-Ribeiro M, Leite-Moreira AF, Pimentel-Nunes P. Role of colonic microbiota in colorectal carcinogenesis: a systematic review. Rev Esp Enferm Dig. 2015;107(11):659–71.PubMed
55.
Zurück zum Zitat Rao M, Gao C, Hou J, Gu J, Law BYK, Xu Y. Non-digestible carbohydrate and the risk of colorectal neoplasia: a systematic review. Nutr Cancer. 2021;73(1):31–44.PubMedCrossRef Rao M, Gao C, Hou J, Gu J, Law BYK, Xu Y. Non-digestible carbohydrate and the risk of colorectal neoplasia: a systematic review. Nutr Cancer. 2021;73(1):31–44.PubMedCrossRef
56.
Zurück zum Zitat Ma Y, Hu M, Zhou L, Ling S, Li Y, Kong B, et al. Dietary fiber intake and risks of proximal and distal colon cancers: a meta-analysis. Medicine (Baltimore). 2018;97(36):e11678.CrossRef Ma Y, Hu M, Zhou L, Ling S, Li Y, Kong B, et al. Dietary fiber intake and risks of proximal and distal colon cancers: a meta-analysis. Medicine (Baltimore). 2018;97(36):e11678.CrossRef
57.
Zurück zum Zitat Sun Q, Jia Q, Song L, Duan L. Alterations in fecal short-chain fatty acids in patients with irritable bowel syndrome: a systematic review and meta-analysis. Medicine (Baltimore). 2019;98(7):e14513.CrossRef Sun Q, Jia Q, Song L, Duan L. Alterations in fecal short-chain fatty acids in patients with irritable bowel syndrome: a systematic review and meta-analysis. Medicine (Baltimore). 2019;98(7):e14513.CrossRef
58.
Zurück zum Zitat Zhuang X, Li T, Li M, Huang S, Qiu Y, Feng R, et al. Systematic review and meta-analysis: short-chain fatty acid characterization in patients with inflammatory bowel disease. Inflamm Bowel Dis. 2019;25(11):1751–63.PubMedCrossRef Zhuang X, Li T, Li M, Huang S, Qiu Y, Feng R, et al. Systematic review and meta-analysis: short-chain fatty acid characterization in patients with inflammatory bowel disease. Inflamm Bowel Dis. 2019;25(11):1751–63.PubMedCrossRef
59.
Zurück zum Zitat Rios-Covian D, González S, Nogacka AM, Arboleya S, Salazar N, Gueimonde M, et al. An overview on fecal branched short-chain fatty acids along human life and as related with body mass index: associated dietary and anthropometric factors. Front Microbiol. 2020;11:973.PubMedPubMedCentralCrossRef Rios-Covian D, González S, Nogacka AM, Arboleya S, Salazar N, Gueimonde M, et al. An overview on fecal branched short-chain fatty acids along human life and as related with body mass index: associated dietary and anthropometric factors. Front Microbiol. 2020;11:973.PubMedPubMedCentralCrossRef
60.
Zurück zum Zitat Gill PA, van Zelm MC, Muir JG, Gibson PR. Review article: short chain fatty acids as potential therapeutic agents in human gastrointestinal and inflammatory disorders. Aliment Pharmacol Ther. 2018;48(1):15–34.PubMedCrossRef Gill PA, van Zelm MC, Muir JG, Gibson PR. Review article: short chain fatty acids as potential therapeutic agents in human gastrointestinal and inflammatory disorders. Aliment Pharmacol Ther. 2018;48(1):15–34.PubMedCrossRef
61.
Zurück zum Zitat Shuwen H, Miao D, Quan Q, Wei W, Zhongshan Z, Chun Z, et al. Protective effect of the “food-microorganism-SCFAs” axis on colorectal cancer: from basic research to practical application. J Cancer Res Clin Oncol. 2019;145(9):2169–97.PubMedCrossRef Shuwen H, Miao D, Quan Q, Wei W, Zhongshan Z, Chun Z, et al. Protective effect of the “food-microorganism-SCFAs” axis on colorectal cancer: from basic research to practical application. J Cancer Res Clin Oncol. 2019;145(9):2169–97.PubMedCrossRef
62.
Zurück zum Zitat Akimoto N, Ugai T, Zhong R, Hamada T, Fujiyoshi K, Giannakis M, et al. Rising incidence of early-onset colorectal cancer - a call to action. Nat Rev Clin Oncol. 2021;18(4):230–43.PubMedCrossRef Akimoto N, Ugai T, Zhong R, Hamada T, Fujiyoshi K, Giannakis M, et al. Rising incidence of early-onset colorectal cancer - a call to action. Nat Rev Clin Oncol. 2021;18(4):230–43.PubMedCrossRef
63.
Zurück zum Zitat Burnett-Hartman AN, Lee JK, Demb J, Gupta S. An update on the epidemiology, molecular characterization, diagnosis, and screening strategies for early-onset colorectal cancer. Gastroenterology. 2021;160(4):1041–9.PubMedCrossRef Burnett-Hartman AN, Lee JK, Demb J, Gupta S. An update on the epidemiology, molecular characterization, diagnosis, and screening strategies for early-onset colorectal cancer. Gastroenterology. 2021;160(4):1041–9.PubMedCrossRef
64.
Zurück zum Zitat Saad El Din K, Loree JM, Sayre EC, Gill S, Brown CJ, Dau H, et al. Trends in the epidemiology of young-onset colorectal cancer: a worldwide systematic review. BMC Cancer. 2020;20(1):288.PubMedCrossRef Saad El Din K, Loree JM, Sayre EC, Gill S, Brown CJ, Dau H, et al. Trends in the epidemiology of young-onset colorectal cancer: a worldwide systematic review. BMC Cancer. 2020;20(1):288.PubMedCrossRef
65.
Zurück zum Zitat Vuik FE, Nieuwenburg SA, Bardou M, Lansdorp-Vogelaar I, Dinis-Ribeiro M, Bento MJ, et al. Increasing incidence of colorectal cancer in young adults in Europe over the last 25 years. Gut. 2019;68(10):1820–6.PubMedCrossRef Vuik FE, Nieuwenburg SA, Bardou M, Lansdorp-Vogelaar I, Dinis-Ribeiro M, Bento MJ, et al. Increasing incidence of colorectal cancer in young adults in Europe over the last 25 years. Gut. 2019;68(10):1820–6.PubMedCrossRef
66.
Zurück zum Zitat Kastrinos F, Samadder NJ, Burt RW. Use of family history and genetic testing to determine risk of colorectal cancer. Gastroenterology. 2020;158(2):389–403.PubMedCrossRef Kastrinos F, Samadder NJ, Burt RW. Use of family history and genetic testing to determine risk of colorectal cancer. Gastroenterology. 2020;158(2):389–403.PubMedCrossRef
70.
Zurück zum Zitat Ocvirk S, Wilson AS, Appolonia CN, Thomas TK, O'Keefe SJD. Fiber, fat, and colorectal cancer: new insight into modifiable dietary risk factors. Curr Gastroenterol Rep. 2019;21(11):62.PubMedCrossRef Ocvirk S, Wilson AS, Appolonia CN, Thomas TK, O'Keefe SJD. Fiber, fat, and colorectal cancer: new insight into modifiable dietary risk factors. Curr Gastroenterol Rep. 2019;21(11):62.PubMedCrossRef
72.
Zurück zum Zitat Jia W, Xie G, Jia W. Bile acid-microbiota crosstalk in gastrointestinal inflammation and carcinogenesis. Nat Rev Gastroenterol Hepatol. 2018;15(2):111–28.PubMedCrossRef Jia W, Xie G, Jia W. Bile acid-microbiota crosstalk in gastrointestinal inflammation and carcinogenesis. Nat Rev Gastroenterol Hepatol. 2018;15(2):111–28.PubMedCrossRef
73.
Zurück zum Zitat Davidson KW, Barry MJ, Mangione CM, Cabana M, Caughey AB, Davis EM, et al. Screening for colorectal cancer: US Preventive Services Task Force Recommendation Statement. Jama. 2021;325(19):1965–77.PubMedCrossRef Davidson KW, Barry MJ, Mangione CM, Cabana M, Caughey AB, Davis EM, et al. Screening for colorectal cancer: US Preventive Services Task Force Recommendation Statement. Jama. 2021;325(19):1965–77.PubMedCrossRef
74.
Zurück zum Zitat Ferrari A, Neefs I, Hoeck S, Peeters M, Van Hal G. Towards novel non-invasive colorectal cancer screening methods: a comprehensive review. Cancers (Basel). 2021;13(8):1820. Ferrari A, Neefs I, Hoeck S, Peeters M, Van Hal G. Towards novel non-invasive colorectal cancer screening methods: a comprehensive review. Cancers (Basel). 2021;13(8):1820.
75.
Zurück zum Zitat Jodal HC, Helsingen LM, Anderson JC, Lytvyn L, Vandvik PO, Emilsson L. Colorectal cancer screening with faecal testing, sigmoidoscopy or colonoscopy: a systematic review and network meta-analysis. BMJ Open. 2019;9(10):e032773.PubMedPubMedCentralCrossRef Jodal HC, Helsingen LM, Anderson JC, Lytvyn L, Vandvik PO, Emilsson L. Colorectal cancer screening with faecal testing, sigmoidoscopy or colonoscopy: a systematic review and network meta-analysis. BMJ Open. 2019;9(10):e032773.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Imperiale TF, Gruber RN, Stump TE, Emmett TW, Monahan PO. Performance characteristics of fecal immunochemical tests for colorectal cancer and advanced adenomatous polyps: a systematic review and meta-analysis. Ann Intern Med. 2019;170(5):319–29.PubMedCrossRef Imperiale TF, Gruber RN, Stump TE, Emmett TW, Monahan PO. Performance characteristics of fecal immunochemical tests for colorectal cancer and advanced adenomatous polyps: a systematic review and meta-analysis. Ann Intern Med. 2019;170(5):319–29.PubMedCrossRef
77.
Zurück zum Zitat Anghel SA, Ionita-Mindrican CB, Luca I, Pop AL. Promising epigenetic biomarkers for the early detection of colorectal cancer: a systematic review. Cancers (Basel). 2021;13(19):4965. Anghel SA, Ionita-Mindrican CB, Luca I, Pop AL. Promising epigenetic biomarkers for the early detection of colorectal cancer: a systematic review. Cancers (Basel). 2021;13(19):4965.
Metadaten
Titel
Short-chain fatty acid concentrations in the incidence and risk-stratification of colorectal cancer: a systematic review and meta-analysis
verfasst von
Ehsan Alvandi
Wilson K. M. Wong
Mugdha V. Joglekar
Kevin J. Spring
Anandwardhan A. Hardikar
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
BMC Medicine / Ausgabe 1/2022
Elektronische ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-022-02529-4

Weitere Artikel der Ausgabe 1/2022

BMC Medicine 1/2022 Zur Ausgabe

Leitlinien kompakt für die Allgemeinmedizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Facharzt-Training Allgemeinmedizin

Die ideale Vorbereitung zur anstehenden Prüfung mit den ersten 24 von 100 klinischen Fallbeispielen verschiedener Themenfelder

Mehr erfahren

Update Allgemeinmedizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.