Next Article in Journal
Response to COVID-19 Vaccination in Patients with Inflammatory Bowel Disease on Biological Treatment
Previous Article in Journal
Different Clinical Features of Idiopathic Achalasia in Various Countries
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Microbiota and the Relationship with Colorectal Cancer: Surgical Complications—A Review

1
Department 8—Radiology, Oncology, Hematology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, Bulevardul Eroii Sanitari 8, 050474 Bucharest, Romania
2
Radiation Therapy Department, “Prof. Dr. Al. Trestioreanu” Oncology Institute, Sos. Fundeni No. 252, 022328 Bucharest, Romania
3
Department 10—General Surgery, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, Bulevardul Eroii Sanitari 8, 050474 Bucharest, Romania
4
Department 3—Physiology, Faculty of Dental Medicine, “Carol Davila” University of Medicine and Pharmacy, Bulevardul Eroii Sanitari 8, 050474 Bucharest, Romania
5
Department 3—Nephrology, Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, Bulevardul Eroii Sanitari 8, 050474 Bucharest, Romania
6
National Institute of Infectious Diseases “Prof. Dr. Matei Bals”, 021105 Bucharest, Romania
*
Author to whom correspondence should be addressed.
Gastrointest. Disord. 2022, 4(2), 66-76; https://0-doi-org.brum.beds.ac.uk/10.3390/gidisord4020008
Submission received: 25 March 2022 / Revised: 1 April 2022 / Accepted: 22 April 2022 / Published: 29 April 2022
(This article belongs to the Special Issue Colorectal Cancer: From Pathophysiology to Novel Therapeutic Approach)

Abstract

:
Colorectal cancer (CRC) is one of the most common cancers and represents a major global health burden. While genetics are implicated in a portion of CRC patients, most cases are sporadic. A new possibility of tumor initiation and promotion might be microbiome composition. It was recently shown that bacteria from the gut microbiome might be used as biomarkers for CRC detection, especially Fusobacterium nucleatum, Peptostreoptococcus stomatis, Parvimonas mica, Solobacterium moorei, and Peptostreptococcus anaerobius. Conversely, the healthy gut microbiome is mostly colonized by Bacterioides (Bacterioides fragilis, vulgatus, uniformis), Firmicutes (Clostridium spp., Ruminococcus faecis, Enterococcus faecium), and Actinobacteria (Bifidobacterium bifidum). Some strains of gut bacteria favor tumor promotion through DNA and RNA damage (directly or through interaction with other known food carcinogens) and through local immune inhibition. It is possible that bacteria (e.g., Bacillus polyfermenticus, Alistipes shahii, Lactobacillus casei) exist with protective functions against tumor promotion. Despite current advances in colorectal cancer treatment, especially in the medical oncology and radiotherapy domains, surgery remains the mainstay of curative treatment for colorectal cancer patients, even in the oligometastatic setting. Surgical complications like anastomotic leakage, excessive blood loss, abscess, and abdominal sepsis can reduce 1-year and 5-year overall survival and increase the recurrence rates for these patients; therefore, we reviewed currently published data focusing on the relationship between gut microbiota and postoperative complications for colorectal cancer patients.

1. Introduction

Colorectal cancer (CRC) is one of the most common cancers and represents a major global health burden. The latest statistics show a worldwide incidence of 1.93 million CRC cases in 2020, with almost one million deaths [1]. While genetics are implicated in a portion of CRC patients, most cases are sporadic. In some situations, well-known risk factors such as high lipid intake, high body mass index, lack of exercise, and alcohol consumption might be present. A series of steps are involved in the process of oncogenesis, as reviewed by Hanahan, including inflammation, microenvironmental changes, and immune inhibition [2]. Moreover, in the context of the intestinal digestive tract, a special role is played by the local flora. Given the presence of the rich intestinal flora, a new possibility for influencing tumor initiation and promotion might be microbiome composition [3,4]. Blood group type can be associated with some malignancies, as proved by the study of Gaube A. et al., while in colorectal cancer, the results reported by Khalili et al. do not support an association between ABO blood group and risk of colorectal cancer [5,6].
There are many findings regarding the microbiome in relation to gut health, inflammatory responses, type 2 diabetes, and even mental health [7,8,9]. The digestive microbiome is composed of millions of microorganisms and each segment of the digestive tract has its own bacterial composition. These organisms obtain nutrients from dietary components, and, in turn, they contribute to the production of gases, acetate, propionate, and butyrate. Most of these bacteria come from the Gram-negative Bacteroidetes and Firmicutes. The latter phylum produces butyrate, which is associated with inflammatory response modulation, has a role in cell proliferation and differentiation, and also has anti-neoplastic functions [10,11]. Other less abundant bacteria such as Bifidobacterium, Escherichia coli, and Akkermansia muciniphila are present and have a larger impact on health. Bifidobacterium breve was shown to decrease the severity of diarrhea, alleviate celiac disease, and even influence the outcomes of neurological disorders and mental disorders [12,13,14]. Akkermansia muciniphila, by producing a thick mucus, helps to form a barrier on the intestinal wall and modulate local immune receptors [15].
Recently, the scientific corpus regarding the gut microbiome and its role in health modulation has consistently increased due to better sequencing algorithms and improved hardware tools such as “bacteria for identification and quantification” [16,17]. A method for microbiome profiling is to use targeted amplicon sequencing. This method uses universal primers for highly conserved DNA such as the 16S ribosomal RNA subunit (rRNA) which is encoded by ribosomal DNA (rDNA). This target is present in all bacteria and, in this method, it is amplified by polymerase chain reaction (PCR). Another method is to use metagenomic sequencing, which involves the analysis of the entire genomes of all bacteria from a given site using a shotgun approach [18,19]. These sequencing techniques create a new challenge relating to the analysis of large data sets. Methods for analysis and visualization such as alpha diversity (maximum species richness) and beta diversity (comparison of bacterial communities) are applied via software solutions such as QIIME, Mothur [20,21].
It is possible that bacteria from the gut microbiome might be used as biomarkers for CRC detection, especially Fusobacterium nucleatum, Peptostreoptococcus stomatis and anaerobius, Parvimonas mica, and Solobacterium moorei [22,23,24,25]. Conversely, the healthy gut microbiota mostly consists of Bacterioides (Bacterioides fragilis, vulgatus, uniformis), Firmicutes (Clostridium spp., Ruminococcus faecis, Enterococcus faecium), and Actinobacteria (Bifidobacterium bifidum). It has been shown that some strains of gut bacteria favor tumor promotion through DNA and RNA damage (directly or through interaction with other known food carcinogens) and through local immune inhibition [26,27,28]. There is also the possibility that protective bacteria (e.g., Bacillus polyfermenticus, Alistipes shahii, Lactobacillus casei) against tumor promotion exist, as reviewed by Zitvogel et al. [29].
Despite current advances in colorectal cancer treatment, especially in the chemotherapy, target therapy, immunotherapy, and radiotherapy treatment modalities, surgery remains the mainstay curative treatment for colorectal cancer patients, even in the oligometastatic setting [30,31,32]. Surgical complications like anastomotic leakage, excessive blood loss, abscess, and abdominal sepsis can reduce 1-year and 5-year overall survival and increase the recurrence rates for these patients [33]; therefore, we reviewed currently published data focusing on the relationship between gut microbiota and postoperative complications for colorectal cancer patients. We have summarized in Table 1 the main postsurgical complications along with the prevalent bacteria involved in these processes and their possible effects.

2. Alterations of Intestinal Microbiota Following Colorectal Surgery

There are several factors that could be involved in changes to the composition of the gut microbiota after colorectal surgery. One factor is stress, related to gut microbiota through the microbiome–gut–brain axis. Stress can influence the composition of the microbiome, and the microbiome can influence the host response to stress through immune, endocrine, and metabolic pathways [34]. Another factor is oxygen exposure during colorectal surgery, which could cause a significant depletion of the facultative anaerobes and obligate anaerobes [34]. In 2014, Shogan et al. demonstrated that in rat models, the opening of the bowel caused a loss of “good” obligate anaerobes, such as Bacteroides, and a gain of “bad” facultative anaerobes, such as Enterococcus [35]. Research on rat models identified that vascular changes secondary to colorectal surgery may cause temporary or permanent ischemia of the neighboring gastrointestinal tract, thus leading to significant changes in the gut microbiota [35]. Using a reperfusion model, the authors found that changes in gut microbiota started to develop 30 min following colic ischemia and reached a peak at 6 h after reperfusion before gradually recovering. This change consisted of an early increase in E. coli and Prevotella and a later increase in Lactobacillus, in line with reperfusion and epithelial healing [36]. Ohigashi et al. conducted the first study to assess the impact of CRC surgery on the fecal microbiota through 16S rRNA-targeted reverse transcription-quantitative PCR (RT-qPCR) analysis. The results demonstrated a decreased abundance in obligate anaerobic bacteria, including members of Bacteroides, Bifidobacterium, Clostridium, and Prevotella, which are crucial for intestinal homeostasis, whereas pathogens such as Enterococcus, Pseudomonas, and Staphylococcus were enriched postoperatively [36].
A decreased diversity in the fecal microbiota, such as a lower ratio of Firmicutes and Bacteroidetes to Proteobacteria and increased levels of Klebsiella, were found in CRC patients after the surgery compared to pre-operative CRC patients or to healthy individuals [37,38]. This result has two facets—first, a higher risk for an inflammatory response was observed postsurgery, and second, the strains that are known to be associated with a higher recurrence risk (Enterococcus, Fusobacterium) were also destroyed [39]. In contrast, a study done by Liu et al. [40] showed that preoperative levels of Escherichia–Shigella genera were higher versus levels in postoperative patients, while postoperative levels of Enterococcus and Parabacteroides were higher.

3. Anastomotic Leakage and Colorectal Microbiota

Anastomotic leakage (AL) is defined as a defect of the intestinal wall at the anastomotic site, leading to communication between the intra- and extraluminal compartments. The severity of anastomotic leakage should be graded according to its impact on clinical management. Grade A anastomotic leakage results in no change in patient management, whereas grade B leakage requires active therapeutic intervention but is manageable without re-laparotomy. Grade C anastomotic leakage requires re-laparotomy [41]. The microbiome is a new and very promising field of research, especially when studying the etiologies of AL in colorectal surgery. Identifying at-risk patients with unfavorable microbiome compositions, comprising pathogens with high collagenase activity, and treating them with appropriate antibiotic regimen (per os, intravenous, or by enema) and/or fecal transplantation if required could help to reduce AL rate [42]. Further, studying the microbiome might help to explain the protective effect of preoperative oral antibiotics on AL rate. Recent evidence supports the hypothesis that AL might result from a local infective complication, resulting in impaired healing at the anastomotic level due to a local increase in collagenase activity [43].
Jasper et al., in a pilot study, collected fecal samples from 16 patients that had C-seal, of which 8 developed AL. Bacterial 16S rDNA of both groups were analyzed using MISeq technique. They found that Lachnospiraceae and Bacteroidaceae families were higher in patients with AL, especially Ruminococcus, Blautia, Roseburia, and Coprococcus. The most important involved bacteria were the mucin-degrading Ruminococci (R. gnavus and R. torques). Another interesting finding in their study related high BMI to the same bacterial family, especially R. obeum, although this was not statistically significant (p = 0.068). These findings were surprising, given that it was shown that butyrate-producing bacteria were associated with reduced oxidative stress and with decreased risk of inflammation [44,45]. Later, the same authors did a more extensive study on 123 patients after surgery, out of which 63 had C-seal. They concluded that AL in patients without C-seal could be explained by microbiome diversity, specifically higher levels of Lachnospiraceae and Bacteroidaceae families [46]. Their findings suggest no association between microbiome and AL in C-seal patients, although the C-seal group had a higher incidence of AL (25% vs. 17%).
In another study by Palmisano et al., 49 stool samples were analyzed from 27 healthy subjects and 22 patients that had surgery for colorectal cancer, out of which 5 developed AL. They found that the second group had significantly higher levels of Bacteroides family bacteria (Bacteroides fragilis and Bacteroides faecis) compared to healthy subjects (1.5% vs. 0.2% and 1.9% vs. 1.3%). Within the second subgroup, microbiome composition was different between patients that developed AL vs. those that did not—there was a higher level of Acinetobacter Iwoffi, johnsonii in AL patients, while other species had higher incidence in non-AL patients—Barnesiella intestinihominis (4%, p < 0.05) and Faecalibacterium prausnitzii (2%) (Table 1) [47]. Another possible association with AL risk was shown for the Bifidobacterium genus (odds radio 3.96, CI = 1.50–10.51) [48]. It was shown that Bifidobacterium inhibits PTGS2 expression, which was shown to promote neovascularization [49,50].
These observations suggest that the C-seal influences the microbial composition after its introduction and that this may ultimately impair anastomotic healing [51]. Moreover, the study of Shogan BD et al. demonstrated that Enterococcus faecalis leads to the degradation of collagen IV at the anastomotic level through activation of matrix metallo-proteinase (MMP)-9. In the same study, topical antibiotherapy administered by enema and targeting Enterococcus faecalis was shown to reduce AL to 0%, whereas intramuscular Cefotixin—commonly used for elective surgery prophylaxis—did not reduce collagenase activity or AL risk [52]. Regarding MMP, in a study from Stumpf et al., a lower collagen type I/III ratio and higher expression of MMP-1, -2, and -9 in biopsies of patients with impaired anastomotic healing was found when compared to controls [53]. All these data suggest that an unfavorable microbiome comprising collagenase-inducing pathogens and neovascularization modulation might impair anastomotic healing and result in AL, and that changes in the local microbiome caused by surgery might worsen the situation of patients with already unfavorable microbiome profiles [54].

4. Infectious Complications

Surgical site infection (SSI) following colorectal surgery is a frequent complication associated with increases in morbidity, medical expenses, and mortality. The estimated incidence rate of SSI following colorectal surgery is currently considered to be up to 20%. The most common causative pathogen is Staphylococcus aureus (S. aureus), which is responsible for 20% of SSIs and, due to high use of antibiotherapy, methicillin-resistant S. aureus [55].
The overall surgical site infection rate was found to be 9.6%, of which 5.5% were incisional surgical site infections and 4.1% were organ/space surgical site infections. Patients with rectal cancer had a two-fold increased risk for surgical site infection compared to colon cancer patients. Surgical site infection was associated with lower disease-free survival. In patients that developed SSI, E. coli and P. aeruginosa were the agents found in patients with colon cancer, while E. coli and Enterococcus spp. were found in patients with rectal cancer [36,56].
In a study by Naoya Aisu et al., 165 CRC patients were divided into two groups, with one group (n = 75) receiving probiotic tablets with Clostridium butyricum, Bacillus mesentericus, and Enterococcus faecalis while the other group did not. Following the rate of surgical site infections, the authors found that in the nonprobiotic group, 16 patients developed SSI compared to only 5 cases in the probiotic group (p = 0.016), thus showing a statistically significant decrease in SSI with probiotic administration [57].
Generally, the interplay between bacterial strains is complicated and the colonization of tissue with some types of strains might inhibit new strains’ development (competitive exclusion) [58]. This seems to be the case for methicillin-resistant staphylococcus aureus (MRSA). Strains of Lactobacillus and Bifidobacterium were found to inhibit MRSA growth in vitro as well as in vivo [59,60]. Liu Guanwen et al. showed that Lactobacillus rhamonsus isolated from human milk inhibited the growth of S. aureus in vitro as well as in animal models through α tumor necrosis factor (TNF-α) and interleukin 6 (IL-6) decrease [61].
One other possible mechanism might involve surgical stress, which could promote contamination of microbiota with skin bacteria and lead to the disruption of the local T lymphocytes, thus creating an increased risk of SSI [56,62,63].

5. Postoperative Ileus

Postoperative ileus (POI) is a temporary gastrointestinal motility disorder characterized by nausea, vomiting, inability to eat, and lack of flatus and stool [64]. It has been found that 10–30% of patients have increased morbidity and prolonged hospitalization due to postoperative ileus.
Serotonin, a key neurotransmitter in the digestive tract neurons, with a role in motility and contractility, is modulated by the components of the microbiome and it might be involved in POI production. In a meta-analysis, Drake et al. concluded that serotonin agonists along with μ-receptor opioid agonists reduced the duration of POI. [65] Moreover, a link between microbiome composition and serotonin modulation exists. This relation is due to the stimulation of the colonic enterochromaffin cells by local flora, which in turn leads to the activation of 5-hydroxytryptamine (5-HT) biosynthesis; some bacteria might even directly produce 5-HT [66,67]. It is possible then that the microbiota influence the length of postoperative ileus.
Another possible factor that might influence POI seems to be the composition of the gut microbiome through the modulation of motility and local inflammation response [68]. One such example comes from a study from Ye Jin et al. in which two patient cohorts that had CRC surgery were analyzed [38]. They found that Firmicutes-to-Bacteroidetes ratio and Fusobacteria were lower in patients with ileus versus those without, while Escherichia–higella, Veillonella, and Ralstonia were present in higher levels in the patients that developed postoperative ileus. Faecalibacterium was also significantly reduced in the former group [69]. The authors introduced a second cohort in their study and used Faecalibacterium levels as a biomarker to predict the development of postoperative ileus. The AUC obtained for the validation cases was 0.79, a finding that could be used to validate other independent cases using the same model.
In another study on rectal cancer patients that had surgery, Shogan et al. found that patients that developed postoperative ileus had increased levels of Proteobacteria and Bacteroidetes and decreased levels of Actinobacteria and Firmicutes. These changes depended on the postoperative day, with abundance increasing with the number of postoperative days. Moreover, they considered possible confounding variables such as previous intestinal surgery, steroid use, and preoperative chemotherapy/radiotherapy and none were found to affect postoperative ileus risk, thus underscoring the role of microbiome composition in this postsurgical complication [70].
In a study on mice, Pohl et al. described a connection between CD103+, CD11b+ cells, and microbiota which might regulate postoperative ileus. A substantial reduction of interleukin-12 (IL-12) and Nitric Oxide Synthase (iNOS) was identified, and this was seen as a contributor to postoperative ileus following gut microbiota depletion with oral antibiotics [71].

6. Postoperative Adhesions

Peritoneal adhesions are pathological bonds that occur usually between the omentum, loops of bowel, and the abdominal wall. They are mostly induced by surgical procedures in the peritoneal cavity, and their prevalence after major abdominal procedures has been evaluated at 63–97%. Colorectal surgery has proven to be the most significant type of surgery that may cause intra-abdominal adhesions [72].
Peritoneal irritation by infection or surgical trauma can induce a local inflammatory response, with an imbalance in fibrin deposition/degradation leading to peritoneal adhesions. More specifically, a factor in response to tissue damage during abdominal surgery is the local migration of fibroblasts, which tends to induce extracellular matrix production. Matrix metalloprotease can reverse this response and normal healing is then reestablished. Disturbance of this balance leads to peritoneal adhesion formation and microbiota might modulate these responses [72]. It was shown that nonsteroidal anti-inflammatory drugs mitigate the inflammation in animal models, although it is not yet clear if these findings are consistent in human patients [73,74].
An experimental study on such modulation was done by Bothin et al., wherein the authors compared germ-free rats (GF) with ex-germ-free rats contaminated with E. coli or Lactobacillus. They found that GF rats had decreased peritoneal adhesion formation compared to rats contaminated with E. coli or Lactobacillus. The authors concluded that the probable bacterial leakage might be the main culprit [75]. These findings are also consistent with other results in animal models in which antibiotherapy was found to neutralize most microbiome composition, leading to a decreased risk of adhesion formation [76]. Other similar important findings showed an association of lower serotonin production with short-chain fatty acid synthesis in GF mice [77,78].

7. Long-Term Outcomes

Patients that have surgery for CRC usually develop complications in the long term, and it has been found that microbiota can also influence these. Two types of bacteria have been shown to be involved in the pathogenesis of new colorectal cancers and adverse outcomes—Fusobacterium nucleatum (F. nucleatum) and Bacteroides fragilis (B. fragilis). It seems that F. nucleatum is associated with an increased risk of malignant transformation, as was shown by Flanagan et al. Enterotoxigenic B. fragilis was found in abundance in patients that had low-grade dysplasia compared to healthy individuals. Lin et al. studied long-term metabolic alterations after CRC surgery. They found that patients with right hemicolectomy had a higher risk of intestinal dysbiosis, as shown by a lower Firmicutes/Bacteroidetes ratio, higher levels of Fusobacterium, and lower levels of Faecalibacterium prausnitzii compared to the patients with lower anterior resection.
In the study by Flanagan and colleagues, shortened recurrence-free survival was demonstrated in CRC patients with higher levels of F. nucleatum. The presence of enterotoxigenic B. fragilis in the colonic mucosa was associated with a higher colorectal cancer stage [79]. Additionally, Wei et al. concluded that the abundance of F. nucleatum or B. fragilis was a prognostic biomarker of poor survival, associated with increased levels of inflammatory mediators including MMP-9. In addition, the authors identified that B. fragilis can induce NF-κB signaling and release of inflammatory cytokines. Thus, one intervention that can shape the microbiome before surgery and potentially downstage colorectal cancer is mechanical and antibiotic bowel preparation [80].
On the other hand, it was shown that Bifidobacterium might stimulate a local antitumor immune response through effects similar to that of programmed cell death protein ligand 1 (PD-L1) or through death protein 1 (PD-1) and enhanced CD8+ T cells, which might offer a more favorable outcome in patients with CRC [81]. These effects on immune modulation might work in synergy with anti-PD-1/PD-L1 monoclonal antibodies given to patients that are PD-L1 positive or who have microsatellite instability [82].
Kosumi et al. showed that the number of Bifidobacterium was related to the number of signet-ring cells, a finding that should be confirmed by future studies. The study showed no association between the number of intratumor bacteria and local immune response. The hypothesis was given that the cancerous cells might utilize the lactic acid and acetate produced by bacteria and, given the new tumoral milieu, the protective effect of Bifidobacterium might be mitigated [46].

8. Conclusions and Future Perspectives

Many important findings are available regarding the gut microbiota in relation to gastrointestinal tract health and even to mental health. To date, some findings have supported the idea that bacteria (e.g., E. faecalis, P. aeruginosa, Lactobacillus, Faecalibacterium, Enterococcus faecalis, and E. coli) might influence postsurgical events such as anastomotic healing, infectious complications, ileus, and adhesion formation, while strains such as Bifidobacterium might be used to improve local immune response in healthy individuals and to prevent future colorectal oncologic events. It seems that the context of bacterial composition is important, as was hypothesized in the study of Kosumi et al. where Bifidobacterium, normally known to provide local antitumor properties, promoted tumor growth in patients with CRC and, possibly, the presence of signet-ring cells.
Regarding the risk of anastomotic leak after colorectal surgery, a lower diversity in microbiota seems to be a prognostic factor for AL but only in patients that have classical surgery. It is important to extend the study on C-seal patients, since in this group no correlation was found with microbiome diversity. On the other hand, intervention studies with antibiotics might shed some light on the causal patterns of some bacterial families and even on the mechanisms involved such as collagen degradation by bacteria. Additionally, given the fact that the increased usage of antibiotics has led to resistant bacterial strain selection, the possibility of colonization exclusion through butyrate-producing bacteria might deserve further investigation.
Postoperative infections decrease survival in patients and antibiotherapy is necessary. It would be interesting to check whether strains that normally inhibit MRSA would also have the same effect in patients that had CRC surgery, thus reducing the rate of MRSA infections. One study indeed showed that administration of perioperative probiotic tablets in patients that underwent CRC surgery was associated with a significantly lower risk of SSI. Given these results, other similar investigations should be pursued, using the same probiotic agents and others as well.
Postoperative ileus, another important postsurgical complication, might play a role in the development of intestinal dysbiosis, which in turn might lead to microbiome compositions compatible with CRC recurrence. As for the other complications, Faecalibacterium levels and Firmicutes-to-Bacteroidetes ratio are associated with this surgical complication. Continuation of this line of research might be necessary to validate the model developed by Ye Jin et al. and to check the role of Faecalibacterium as a biomarker for postoperative ileus. Another possibility would be to establish a potential causation relationship by manipulating microbiome composition before and after surgery, for example using either antibiotics or fecal transplant. Along these lines, basic research on the local immune response, as was done by Pohl et al., remains continuously important. Given the immune interplay between bacteria and intestinal mucosa shown by Pohl, the findings of Shogan et al. in which corticosteroids did not seem to influence the risk of postoperative ileus might offer another possibility for investigation. This relationship might be tested using Faecalibacterium levels as well as Firmicutes-to-Bacteroidetes ratio as parameters. Another lead might be to determine the role of serotonin in patients with postoperative ileus. Some studies have demonstrated that serotonin agonists decrease the length of the complication; thus, a study using colonization with serotonin-stimulating bacteria or even with serotonin-producing bacteria might be conducted in animal models.
Postoperative adhesion formation, which poses a risk especially for long-term survivors, is modulated by local inflammatory response. To mitigate the risk, NSAIDs might be used, or other drugs that inhibit neovascularization and fibroblast local invasion. Since metalloproteases degrade the extracellular matrix resulting from fibroblasts, some strains of bacteria might regulate this process, as was shown Wei et al. A possible direction for investigation would be to check whether bacteria such as F. nucleatum or B. fragilis, which are known to modulate MMPs, play a role in this complication. Similarly, buryrate-producing bacteria such as Faecalibacterium prausnitzii are known to decrease the inflammatory response and thus might be another avenue for research in conjunction with postoperative adhesion formation. Antibiotherapy effects on the microbiome in this setting could also be investigated, as it was found in animal models that the onset of adhesion formation was slowed.
These findings are mostly from studies involving animal models, and more studies in human patients are needed to establish causative links between different bacterial strains and postsurgical complications.
A great challenge is represented by the amount of data collected from fecal samples, with this kind of analysis requiring expertise and multidisciplinary collaboration between clinical researchers, statisticians, and bioinformaticians. While some associations might be found between composition and a given risk, one needs to consider possible mechanisms for these effects, construct a physio-pathological model, and add in possible confounding variables such as smoking, diabetes, age and frailty, obesity, and medication.
Studies on human patients still cannot directly point to the direction of causation; all these findings seem only to scratch the surface of a vast domain.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Xi, Y.; Xu, P. Global colorectal cancer burden in 2020 and projections to 2040. Transl. Oncol. 2021, 14, 101174. [Google Scholar] [CrossRef] [PubMed]
  2. Hanahan, D. Hallmarks of cancer: New dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  3. Cheng, Y.; Ling, Z.; Li, L. The intestinal microbiota and colorectal cancer. Front. Immunol. 2020, 11, 3100. [Google Scholar] [CrossRef] [PubMed]
  4. Hislop, G. Trends and risk factors for colorectal cancer. Br. Columbia Med. J. 2022, 42, 131–135. [Google Scholar]
  5. Alexandra, G.; Alexandru, M.; Stefan, C.F.; Petruta-Maria, D.; Gabriel, B.M.; Dragos-Eugen, G.; Teodor, G.M. Blood group type association with head and neck cancer. Hematol. Rep. 2022, 14, 24–30. [Google Scholar] [CrossRef]
  6. Khalili, H.; Wolpin, B.M.; Huang, E.S.; Giovannucci, E.L.; Kraft, P.; Fuchs, C.S.; Chan, A.T. ABO blood group and risk of colorectal cancer. Cancer Epidemiol. Biomark. Prev. 2011, 20, 1017–1020. [Google Scholar] [CrossRef] [Green Version]
  7. Malan-Muller, S.; Valles-Colomer, M.; Raes, J.; Lowry, C.A.; Seedat, S.; Hemmings, S.M.J. The Gut microbiome and mental health: Implications for anxiety- and trauma-related disorders. Omics. J. Integr. Biol. 2018, 22, 90–107. [Google Scholar] [CrossRef]
  8. Wen, L.; Duffy, A. Factors influencing the gut microbiota, inflammation, and type 2 diabetes. J. Nutr. 2017, 147, 1468S–1475S. [Google Scholar] [CrossRef] [Green Version]
  9. Järbrink-Sehgal, E.; Andreasson, A. The gut microbiota and mental health in adults. Curr. Opin. Neurobiol. 2020, 62, 102–114. [Google Scholar] [CrossRef]
  10. Graf, D.; Di Cagno, R.; Fåk, F.; Flint, H.J.; Nyman, M.; Saarela, M.; Watzl, B. Contribution of diet to the composition of the human gut microbiota. Microb. Ecol. Health Dis. Taylor Fr. 2015, 26, 26164. [Google Scholar] [CrossRef]
  11. Bach Knudsen, K.E.; Lærke, H.N.; Hedemann, M.S.; Nielsen, T.S.; Ingerslev, A.K.; Gundelund Nielsen, D.S.; Theil, P.K.; Purup, S.; Hald, S.; Schioldan, A.G.; et al. Impact of diet-modulated butyrate production on intestinal barrier function and inflammation. Nutrients 2018, 10, 1499. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Bozzi Cionci, N.; Baffoni, L.; Gaggìa, F.; Di Gioia, D. Therapeutic microbiology: The role of bifidobacterium breve as food supplement for the prevention/treatment of paediatric diseases. Nutrients 2018, 10, 1723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Kazem, Y.I.; Mahmoud, M.H.; Essa, H.A.; Azmy, O.; Kandeel, W.A.; Al-Moghazy, M.; El-Attar, I.; Hasheesh, A.; Mehanna, N.S. Role of Bifidobacterium spp. intake in improving depressive mood and well-being and its link to kynurenine blood level: An interventional study. J. Complement. Integr. Med. 2021. Available online: https://0-www-degruyter-com.brum.beds.ac.uk/document/doi/10.1515/jcim-2021-0351/html (accessed on 2 February 2022). [CrossRef] [PubMed]
  14. Golfetto, L.; de Senna, F.D.; Hermes, J.; Beserra, B.T.S.; França, F.D.S.; Martinello, F. Lower bifidobacteria counts in adult patients with celiac disease on a gluten-free diet. Arq. Gastroenterol. 2014, 51, 139–143. [Google Scholar] [CrossRef]
  15. Ottman, N.; Geerlings, S.Y.; Aalvink, S.; de Vos, W.M.; Belzer, C. Action and function of Akkermansia muciniphila in microbiome ecology, health and disease. Best Pract. Res. Clin. Gastroenterol. 2017, 31, 637–642. [Google Scholar] [CrossRef] [Green Version]
  16. Gloux, K.; Leclerc, M.; Iliozer, H.; L’Haridon, R.; Manichanh, C.; Corthier, G.; Nalin, R.; Blottière, H.M.; Doré, J. Development of high-throughput phenotyping of metagenomic clones from the human gut microbiome for modulation of eukaryotic cell growth. Appl. Environ. Microbiol. Am. Soc. Microbiol. 2007, 73, 3734–3737. [Google Scholar] [CrossRef] [Green Version]
  17. Jin, J.; Yamamoto, R.; Takeuchi, T.; Cui, G.; Miyauchi, E.; Hojo, N.; Ikuta, K.; Ohno, H.; Shiroguchi, K. High-throughput identification and quantification of single bacterial cells in the microbiota. Nat. Commun. 2022, 13, 863. [Google Scholar] [CrossRef]
  18. Davidson, R.M.; Epperson, L.E. Microbiome sequencing methods for studying human diseases. In Disease Gene Identification; DiStefano, J.K., Ed.; Springer: New York, NY, USA, 2018; pp. 77–90. [Google Scholar] [CrossRef]
  19. Ghurye, J.S.; Cepeda-Espinoza, V.; Pop, M. Metagenomic assembly: Overview, challenges and applications. Yale J. Biol. Med. 2016, 89, 353–362. [Google Scholar]
  20. Bolyen, E.; Rideout, J.R.; Dillon, M.R.; Bokulich, N.A.; Abnet, C.C.; Al-Ghalith, G.A.; Alexander, H.; Alm, E.J.; Arumugam, M.; Asnicar, F.; et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 2019, 37, 852–857. [Google Scholar] [CrossRef]
  21. Schloss, P.D.; Westcott, S.L.; Ryabin, T.; Hall, J.R.; Hartmann, M.; Hollister, E.B.; Lesniewski, R.A.; Weber, C.F. Introducing mothur: Open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl. Environ. Microbiol. 2009, 75, 7537–7541. [Google Scholar] [CrossRef] [Green Version]
  22. Long, X.; Wong, C.C.; Tong, L.; Chu, E.S.H.; Ho Szeto, C.; Go, M.Y.Y.; Coker, O.O.; Chan, A.W.H.; Chan, F.K.L.; Sung, J.J.Y.; et al. Peptostreptococcus anaerobius promotes colorectal carcinogenesis and modulates tumour immunity. Nat. Microbiol. 2019, 4, 2319–2330. [Google Scholar] [CrossRef] [PubMed]
  23. Baxter, N.T.; Ruffin, M.T.; Rogers, M.A.M.; Schloss, P.D. Microbiota-based model improves the sensitivity of fecal immunochemical test for detecting colonic lesions. Genome Med. 2016, 8, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Castellarin, M.; Warren, R.L.; Freeman, J.D.; Dreolini, L.; Krzywinski, M.; Strauss, J.; Barnes, R.; Watson, P.; Allen-Vercoe, E.; Moore, R.A.; et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012, 22, 299–306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Yu, J.; Feng, Q.; Wong, S.H.; Zhang, D.; Liang, Q.Y.; Qin, Y.; Tang, L.; Zhao, H.; Stenvang, J.; Li, Y.; et al. Metagenomic analysis of faecal microbiome as a tool towards targeted non-invasive biomarkers for colorectal cancer. Gut 2017, 66, 70–78. [Google Scholar] [CrossRef] [PubMed]
  26. Yao, Q.; Tang, M.; Zeng, L.; Chu, Z.; Sheng, H.; Zhang, Y.; Zhou, Y.; Zhang, H.; Jiang, H.; Ye, M. Potential of fecal microbiota for detection and postoperative surveillance of colorectal cancer. BMC Microbiol. 2021, 21, 156. [Google Scholar] [CrossRef] [PubMed]
  27. Alhinai, E.A.; Walton, G.E.; Commane, D.M. The role of the gut microbiota in colorectal cancer causation. Int. J. Mol. Sci. 2019, 20, 5295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Belkaid, Y.; Hand, T. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [Green Version]
  29. Zitvogel, L.; Daillère, R.; Roberti, M.P.; Routy, B.; Kroemer, G. Anticancer effects of the microbiome and its products. Nat. Rev. Microbiol. 2017, 15, 465–478. [Google Scholar] [CrossRef]
  30. Georgescu, M.-T.; Patrascu, T.; Serbanescu, L.G.; Anghel, R.M.; Gales, L.N.; Georgescu, F.T.; Mitrica, R.I.; Georgescu, D.E. When should we expect curative results of neoadjuvant treatment in locally advanced rectal cancer patients? Chirurgia 2021, 116, 16–23. [Google Scholar] [CrossRef]
  31. Georgescu, D.E.; Patrascu, T.; Georgescu, T.F.; Tulin, A.; Mosoia, L.; Bacalbasa, N.; Stiru, O.; Georgescu, M.-T. Diabetes mellitus as a prognostic factor for locally advanced rectal cancer. Vivo 2021, 35, 2495–2501. [Google Scholar] [CrossRef]
  32. Kuipers, E.J.; Grady, W.M.; Lieberman, D.; Seufferlein, T.; Sung, J.J.; Boelens, P.G.; van de Velde, C.J.H.; Watanabe, T. Colorectal cancer. Nat. Rev. Dis. Primer 2015, 1, 15065. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Breugom, A.J.; van Dongen, D.T.; Bastiaannet, E.; Dekker, F.W.; van der Geest, L.G.M.; Liefers, G.J.; Marinelli, A.W.K.S.; Mesker, W.E.; Portielje, J.E.A.; Steup, W.H.; et al. Association between the most frequent complications after surgery for stage I-III colon cancer and short-term survival, long-term survival, and recurrences. Ann. Surg. Oncol. 2016, 23, 2858–2865. [Google Scholar] [CrossRef] [PubMed]
  34. Wang, F.; Li, Q.; Wang, C.; Tang, C.; Li, J. Dynamic alteration of the colonic microbiota in intestinal ischemia-reperfusion injury. PLoS ONE 2012, 7, e42027. [Google Scholar] [CrossRef] [PubMed]
  35. Shogan, B.D.; Smith, D.P.; Christley, S.; Gilbert, J.A.; Zaborina, O.; Alverdy, J.C. Intestinal anastomotic injury alters spatially defined microbiome composition and function. Microbiome 2014, 2, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Ohigashi, S.; Sudo, K.; Kobayashi, D.; Takahashi, T.; Nomoto, K.; Onodera, H. Significant changes in the intestinal environment after surgery in patients with colorectal cancer. J. Gastrointest. Surg. Off. J. Soc. Surg. Aliment. Tract. 2013, 17, 1657–1664. [Google Scholar] [CrossRef]
  37. Cong, J.; Zhu, H.; Liu, D.; Li, T.; Zhang, C.; Zhu, J.; Lv, H.; Liu, K.; Hao, C.; Tian, Z.; et al. A pilot study: Changes of gut microbiota in post-surgery colorectal cancer patients. Front. Microbiol. 2018, 9, 2777. [Google Scholar] [CrossRef]
  38. Deng, X.; Li, Z.; Li, G.; Li, B.; Jin, X.; Lyu, G. Comparison of microbiota in patients treated by surgery or chemotherapy by 16S rRNA sequencing reveals potential biomarkers for colorectal cancer therapy. Front. Microbiol. 2018, 9, 1607. [Google Scholar] [CrossRef]
  39. Kong, C.; Gao, R.; Yan, X.; Huang, L.; He, J.; Li, H.; You, J.; Qin, H. Alterations in intestinal microbiota of colorectal cancer patients receiving radical surgery combined with adjuvant CapeOx therapy. Sci. China Life Sci. 2019, 62, 1178–1193. [Google Scholar] [CrossRef]
  40. Liu, C.-J.; Zhang, Y.-L.; Shang, Y.; Wu, B.; Yang, E.; Luo, Y.-Y.; Li, X.-R. Intestinal bacteria detected in cancer and adjacent tissue from patients with colorectal cancer. Oncol Lett. 2019, 17, 1115–1127. [Google Scholar] [CrossRef] [Green Version]
  41. Rahbari, N.N.; Weitz, J.; Hohenberger, W.; Heald, R.J.; Moran, B.; Ulrich, A.; Holm, T.; Wong, W.D.; Tiret, E.; Moriya, Y.; et al. Definition and grading of anastomotic leakage following anterior resection of the rectum: A proposal by the International Study Group of Rectal Cancer. Surgery 2010, 147, 339–351. [Google Scholar] [CrossRef]
  42. Fecal Microbiota Transplantation (FMT) Treatment at IPPM Clinic. 2022. Available online: https://www.ippmclinic.com/en/fecal-transplantation (accessed on 10 February 2022).
  43. Meyer, J.; Naiken, S.; Christou, N.; Liot, E.; Toso, C.; Buchs, N.C.; Ris, F. Reducing anastomotic leak in colorectal surgery: The old dogmas and the new challenges. World J. Gastroenterol. 2019, 25, 5017–5025. [Google Scholar] [CrossRef] [PubMed]
  44. Van Praagh, J.B.; de Goffau, M.C.; Bakker, I.S.; Harmsen, H.J.M.; Olinga, P.; Havenga, K. Intestinal microbiota and anastomotic leakage of stapled colorectal anastomoses: A pilot study. Surg. Endosc. 2016, 30, 2259–2265. [Google Scholar] [CrossRef] [PubMed]
  45. Hamer, H.M.; Jonkers, D.M.A.E.; Bast, A.; Vanhoutvin, S.A.L.W.; Fischer, M.A.J.G.; Kodde, A.; Troost, F.J.; Venema, K.; Brummer, R.-J.M. Butyrate modulates oxidative stress in the colonic mucosa of healthy humans. Clin. Nutr. 2009, 28, 88–93. [Google Scholar] [CrossRef] [PubMed]
  46. Van Praagh, J.B.; de Goffau, M.C.; Bakker, I.S.; van Goor, H.; Harmsen, H.J.M.; Olinga, P.; Klaas, H. Mucus microbiome of anastomotic tissue during surgery has predictive value for colorectal anastomotic leakage. Ann. Surg. 2019, 269, 911–916. [Google Scholar] [CrossRef] [PubMed]
  47. Palmisano, S.; Campisciano, G.; Iacuzzo, C.; Bonadio, L.; Zucca, A.; Cosola, D.; Comar, M.; de Manzini, N. Role of preoperative gut microbiota on colorectal anastomotic leakage: Preliminary results. Updat. Surg. 2020, 72, 1013–1022. [Google Scholar] [CrossRef]
  48. Mima, K.; Sakamoto, Y.; Kosumi, K.; Ogata, Y.; Miyake, K.; Hiyoshi, Y.; Ishimoto, T.; Iwatsuki, M.; Baba, Y.; Iwagami, S.; et al. Mucosal cancer-associated microbes and anastomotic leakage after resection of colorectal carcinoma. Surg. Oncol. 2020, 32, 63–68. [Google Scholar] [CrossRef] [PubMed]
  49. Nurmi, J.T.; Puolakkainen, P.A.; Rautonen, N.E. Bifidobacterium lactis sp. 420 up-regulates cyclooxygenase (cox)-1 and down-regulates cox-2 gene expression in a caco-2 cell culture model. Nutr. Cancer. 2005, 51, 83–92. [Google Scholar] [CrossRef]
  50. Reisinger, K.W.; Schellekens, D.H.S.M.; Bosmans, J.W.A.M.; Boonen, B.; Hulsewé, K.W.E.; Sastrowijoto, P.; Joep, D.; Joep, G.; Martijn, P. Cyclooxygenase-2 is essential for colorectal anastomotic healing. Ann. Surg. 2017, 265, 547–554. [Google Scholar] [CrossRef]
  51. Shogan, B.D.; Carlisle, E.M.; Alverdy, J.C.; Umanskiy, K. Do we really know why colorectal anastomoses leak? J. Gastrointest. Surg. Off. J. Soc. Surg. Aliment. Tract. 2013, 17, 1698–1707. [Google Scholar] [CrossRef]
  52. Shogan, B.D.; Belogortseva, N.; Luong, P.M.; Zaborin, A.; Lax, S.; Bethel, C.; Ward, M.; Muldoon, J.P.; Singer, M. Collagen degradation and MMP9 activation by Enterococcus faecalis contributes to intestinal anastomotic leak. Sci. Transl. Med. 2015, 7, 286ra68. [Google Scholar] [CrossRef] [Green Version]
  53. Stumpf, M.; Klinge, U.; Wilms, A.; Zabrocki, R.; Rosch, R.; Junge, K.; Krones, C.; Schumpelick, V. Changes of the extracellular matrix as a risk factor for anastomotic leakage after large bowel surgery. Surgery 2005, 137, 229–234. [Google Scholar] [CrossRef] [PubMed]
  54. Dubinsky-Pertzov, B.; Temkin, E.; Harbarth, S.; Fankhauser-Rodriguez, C.; Carevic, B.; Radovanovic, I.; Ris, F.; Kariv, Y.; Buchs, N.C.; Schiffer, E.; et al. Carriage of extended-spectrum beta-lactamase-producing Enterobacteriaceae and the risk of surgical site infection after colorectal surgery: A prospective cohort study. Clin. Infect. Dis. 2019, 68, 1699–1704. [Google Scholar] [CrossRef] [PubMed]
  55. Bhattacharya, S.; Pal, K.; Jain, S.; Chatterjee, S.S.; Konar, J. Surgical site infection by methicillin resistant staphylococcus aureus-on decline? J. Clin. Diagn. Res. 2016, 10, DC32. [Google Scholar] [CrossRef] [PubMed]
  56. Huh, J.W.; Lee, W.Y.; Park, Y.A.; Cho, Y.B.; Kim, H.C.; Yun, S.H.; Chun, H.K. Oncological outcome of surgical site infection after colorectal cancer surgery. Int. J. Colorectal. Dis. 2019, 34, 277–283. [Google Scholar] [CrossRef] [PubMed]
  57. Aisu, N.; Tanimura, S.; Yamashita, Y.; Yamashita, K.; Maki, K.; Yoshida, Y.; Sasaki, T.; Takeno, S.; Hoshino, S. Impact of perioperative probiotic treatment for surgical site infections in patients with colorectal cancer. Exp. Ther. Med. 2015, 10, 966–972. [Google Scholar] [CrossRef] [Green Version]
  58. Alverdy, J.C.; Hyoju, S.K.; Weigerinck, M.; Gilbert, J.A. The gut microbiome and the mechanism of surgical infection. Br. J. Surg. 2017, 104, e14–e23. [Google Scholar] [CrossRef] [Green Version]
  59. Sikorska, H.; Smoragiewicz, W. Role of probiotics in the prevention and treatment of meticillin-resistant Staphylococcus aureus infections. Int. J. Antimicrob. Agents 2013, 42, 475–481. [Google Scholar] [CrossRef]
  60. Vesterlund, S.; Karp, M.; Salminen, S.; Ouwehand, A.C.Y. Staphylococcus aureus adheres to human intestinal mucus but can be displaced by certain lactic acid bacteria. Microbiology 2006, 152, 1819–1826. [Google Scholar] [CrossRef] [Green Version]
  61. Liu, G.; Pang, B.; Li, N.; Jin, H.; Li, J.; Wu, W.; Ai, C.; Jiang, C.; Shi, J. Therapeutic effect of Lactobacillus rhamnosus SHA113 on intestinal infection by multi-drug-resistant Staphylococcus aureus and its underlying mechanisms. Food Funct. 2020, 11, 6226–6239. [Google Scholar] [CrossRef]
  62. Poutahidis, T.; Kearney, S.M.; Levkovich, T.; Qi, P.; Varian, B.J.; Lakritz, J.R.; Ibrahim, Y.M.; Chatzigiagkos, A.; Alm, E.J.; Alm, S.E. Microbial symbionts accelerate wound healing via the neuropeptide hormone oxytocin. PLoS ONE 2013, 8, e78898. [Google Scholar] [CrossRef] [Green Version]
  63. Alexandru, M.; Rodica, A.; Dragos-Eugen, G.; Mihai-Teodor, G. Assessing the spleen as an organ at risk in radiation therapy and its relationship with radiation-induced lymphopenia: A retrospective study and literature review. Adv. Radiat. Oncol. 2021, 6, 100761. [Google Scholar] [CrossRef] [PubMed]
  64. Vather, R.; Trivedi, S.; Bissett, I. Defining postoperative ileus: Results of a systematic review and global survey. J. Gastrointest. Surg. 2013, 17, 962–972. [Google Scholar] [CrossRef] [PubMed]
  65. Drake, T.M.; Ward, A.E. Pharmacological management to prevent ileus in major abdominal surgery: A systematic review and meta-analysis. J. Gastrointest. Surg. 2016, 20, 1253–1264. [Google Scholar] [CrossRef] [PubMed]
  66. Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [Green Version]
  67. De Vadder, F.; Grasset, E.; Mannerås Holm, L.; Karsenty, G.; Macpherson, A.J.; Olofsson, L.E.; Bäckhed, F. Gut microbiota regulates maturation of the adult enteric nervous system via enteric serotonin networks. Proc. Natl. Acad. Sci. USA 2018, 115, 6458–6463. [Google Scholar] [CrossRef] [Green Version]
  68. Harnsberger, C.R.; Maykel, J.A.; Alavi, K. Postoperative ileus. Clin. Colon. Rectal. Surg. 2019, 32, 166–170. [Google Scholar] [CrossRef]
  69. Jin, Y.; Geng, R.; Liu, Y.; Liu, L.; Jin, X.; Zhao, F.; Feng, J.; Wei, Y. Prediction of postoperative ileus in patients with colorectal cancer by preoperative gut microbiota. Front. Oncol. 2020, 10, 526009. [Google Scholar] [CrossRef]
  70. Shogan, B.D.; Chen, J.; Duchalais, E.; Collins, D.; Chang, M.; Krull, K.; Krezalek, M.A.; Larson, D.W.; Walther-Antonio, M.R.; Chia, N.; et al. Alterations of the rectal microbiome are associated with the development of postoperative ileus in patients undergoing colorectal surgery. J. Gastrointest. Surg. 2020, 24, 1663–1672. [Google Scholar] [CrossRef]
  71. Pohl, J.-M.; Gutweiler, S.; Thiebes, S.; Volke, J.K.; Klein-Hitpass, L.; Zwanziger, D.; Gunzer, M.; Jung, S.; Agace, W.W.; Kurts, C.; et al. Irf4-dependent CD103+CD11b+ dendritic cells and the intestinal microbiome regulate monocyte and macrophage activation and intestinal peristalsis in postoperative ileus. Gut 2017, 66, 2110–2120. [Google Scholar] [CrossRef] [Green Version]
  72. Arung, W.; Meurisse, M.; Detry, O. Pathophysiology and prevention of postoperative peritoneal adhesions. World J. Gastroenterol. 2011, 17, 4545–4553. [Google Scholar] [CrossRef]
  73. Rodgers, K.E.; Girgis, W.; Campeau, J.D.; di Zerega, G.S. Reduction of adhesion formation by intraperitoneal administration of anti-inflammatory peptide 2. J. Investig. Surg. Off. J. Acad. Surg. Res. 1997, 10, 31–36. [Google Scholar] [CrossRef] [PubMed]
  74. Siegler, A.M.; Kontopoulos, V.; Wang, C.F. Prevention of postoperative adhesions in rabbits with ibuprofen, a nonsteroidal anti-inflammatory agent. Fertil. Steril. 1980, 34, 46–49. [Google Scholar] [CrossRef]
  75. Bothin, C.; Okada, M.; Midtvedt, T.; Perbeck, L. The intestinal flora influences adhesion formation around surgical anastomoses. Br. J. Surg. 2001, 88, 143–145. [Google Scholar] [CrossRef] [PubMed]
  76. Oncel, M.; Kurt, N.; Remzi, F.H.; Sensu, S.S.; Vural, S.; Gezen, C.F.; Cincin, T.G.; Olcay, E. The effectiveness of systemic antibiotics in preventing postoperative, intraabdominal adhesions in an animal model. J. Surg. Res. 2001, 101, 52–55. [Google Scholar] [CrossRef]
  77. Reigstad, C.S.; Salmonson, C.E.; Rainey, J.F.; Szurszewski, J.H.; Linden, D.R.; Sonnenburg, J.L.; Farrugia, G.; Kashyap, P.C. Gut microbes promote colonic serotonin production through an effect of short-chain fatty acids on enterochromaffin cells. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2015, 29, 1395–1403. [Google Scholar] [CrossRef] [Green Version]
  78. Hata, T.; Asano, Y.; Yoshihara, K.; Kimura-Todani, T.; Miyata, N.; Zhang, X.-T.; Takakura, S.; Aiba, Y.; Koga, Y.; Sudo, N. Regulation of gut luminal serotonin by commensal microbiota in mice. PLoS ONE 2017, 12, e0180745. [Google Scholar] [CrossRef]
  79. Flanagan, L.; Schmid, J.; Ebert, M.; Soucek, P.; Kunicka, T.; Liska, V.; Bruha, J.; Neary, P.; Dezeeuw, N.; Tommasino, M.; et al. Fusobacterium nucleatum associates with stages of colorectal neoplasia development, colorectal cancer and disease outcome. Eur. J. Clin. Microbiol. Infect. Dis. Off. Publ. Eur. Soc. Clin. Microbiol. 2014, 33, 1381–1390. [Google Scholar] [CrossRef]
  80. Wei, Z.; Cao, S.; Liu, S.; Yao, Z.; Sun, T.; Li, Y.; Li, J.; Zhang, D.; Zhou, Y. Could gut microbiota serve as prognostic biomarker associated with colorectal cancer patients’ survival? A pilot study on relevant mechanism. Oncotarget 2016, 7, 46158–46172. [Google Scholar] [CrossRef] [Green Version]
  81. Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.-L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti–PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef] [Green Version]
  82. Ntomi, V.; Foukas, P.; Papaconstantinou, D.; Antonopoulou, I.; Pikoulis, A.; Panagiotides, I.; Pikoulis, E.; Syrigos, K. The clinical significance of PD-L1 in colorectal cancer (Review). Oncol. Rep. 2021, 45, 92. [Google Scholar] [CrossRef]
Table 1. Summary of prevalent bacteria found in association with main postsurgical complications. Impact Negative—the microbial agents are associated with increased risk for postsurgical complication. Impact Positive—the microbial agents are associated with decreased risk of postsurgical complication.
Table 1. Summary of prevalent bacteria found in association with main postsurgical complications. Impact Negative—the microbial agents are associated with increased risk for postsurgical complication. Impact Positive—the microbial agents are associated with decreased risk of postsurgical complication.
Postsurgical ComplicationMicrobial AgentsImpact
Anastomotic leakage (AL)Ruminococcus, Blautia, Roseburia, Coprococcus, Acinetobacter Iwoffi, johnsoniiNegative
Bacteroidaceae and Lachnospiraceae familiesNegative without C-seal
Faecalibacterium prausnitziiPositive
Enterococcus faecalisPositive
Infectious complicationsClostridium butyricum, Bacillus mesentericus, Enterococcus faecalisPositive
E. coli, P. aeruginosa, Enterococcus spp.Negative
Postoperative ileusE. coli, Veillonella, Rastonia, Proteobaceria, BacteroidetesNegative
Faecalibacterium,
Actinobacteria, Firmicutes
Positive
Postoperative adhesionsE. coli, LactobacillusNegative
Malignant transformationFusobacterium nucleatum, Bacteroides fragilisNegative
BifidobacteriumPositive
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Michire, A.; Anghel, R.; Draghia, P.M.; Burlacu, M.G.; Georgescu, T.F.; Georgescu, D.E.; Balcangiu-Stroescu, A.-E.; Vacaroiu, I.A.; Barbu, M.; Gaube, A. The Microbiota and the Relationship with Colorectal Cancer: Surgical Complications—A Review. Gastrointest. Disord. 2022, 4, 66-76. https://0-doi-org.brum.beds.ac.uk/10.3390/gidisord4020008

AMA Style

Michire A, Anghel R, Draghia PM, Burlacu MG, Georgescu TF, Georgescu DE, Balcangiu-Stroescu A-E, Vacaroiu IA, Barbu M, Gaube A. The Microbiota and the Relationship with Colorectal Cancer: Surgical Complications—A Review. Gastrointestinal Disorders. 2022; 4(2):66-76. https://0-doi-org.brum.beds.ac.uk/10.3390/gidisord4020008

Chicago/Turabian Style

Michire, Alexandru, Rodica Anghel, Petruta Maria Draghia, Mihnea Gabriel Burlacu, Teodor Florin Georgescu, Dragos Eugen Georgescu, Andra-Elena Balcangiu-Stroescu, Ileana Adela Vacaroiu, Maria Barbu, and Alexandra Gaube. 2022. "The Microbiota and the Relationship with Colorectal Cancer: Surgical Complications—A Review" Gastrointestinal Disorders 4, no. 2: 66-76. https://0-doi-org.brum.beds.ac.uk/10.3390/gidisord4020008

Article Metrics

Back to TopTop