Next Article in Journal
Core Endophytic Bacteria and Their Roles in the Coralloid Roots of Cultivated Cycas revoluta (Cycadaceae)
Next Article in Special Issue
The Contribution of the Intestinal Microbiota to the Celiac Disease Pathogenesis along with the Effectiveness of Probiotic Therapy
Previous Article in Journal
Transcriptome and Metabolome Analyses of Thitarodes xiaojinensis in Response to Ophiocordyceps sinensis Infection
Previous Article in Special Issue
Old Folks, Bad Boon: Antimicrobial Resistance in the Infant Gut Microbiome
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut Microbiota in Diagnosis, Therapy and Prognosis of Cholangiocarcinoma and Gallbladder Carcinoma—A Scoping Review

1
Department of General, Visceral and Transplant Surgery, University Medical Center, Johannes Gutenberg University, 55131 Mainz, Germany
2
Center for Complementary Medicine, Department of Medicine II, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
3
Institute of Medical Biometry and Statistics (IMBI), Faculty of Medicine and Medical Center, University of Freiburg, 79104 Freiburg, Germany
4
Institute of Medical Microbiology and Hygiene, Medical Center—University of Freiburg, Faculty of Medicine, University of Freiburg, 79104 Freiburg, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Microorganisms 2023, 11(9), 2363; https://doi.org/10.3390/microorganisms11092363
Submission received: 20 July 2023 / Revised: 9 September 2023 / Accepted: 19 September 2023 / Published: 21 September 2023
(This article belongs to the Special Issue Latest Review Papers in Gut Microbiota 2023)

Abstract

:
Cancers of the biliary tract are more common in Asia than in Europe, but are highly lethal due to delayed diagnosis and aggressive tumor biology. Since the biliary tract is in direct contact with the gut via the enterohepatic circulation, this suggests a potential role of gut microbiota, but to date, the role of gut microbiota in biliary tract cancers has not been elucidated. This scoping review compiles recent data on the associations between the gut microbiota and diagnosis, progression and prognosis of biliary tract cancer patients. Systematic review of the literature yielded 154 results, of which 12 studies and one systematic review were eligible for evaluation. The analyses of microbiota diversity indices were inconsistent across the included studies. In-depth analyses revealed differences between gut microbiota of biliary tract cancer patients and healthy controls, but without a clear tendency towards particular species in the studies. Additionally, most of the studies showed methodological flaws, for example non-controlling of factors that affect gut microbiota. At the current stage, there is a lack of evidence to support a general utility of gut microbiota diagnostics in biliary tract cancers. Therefore, no recommendation can be made at this time to include gut microbiota analyses in the management of biliary tract cancer patients.

1. Introduction

Biliary tract cancer (BTC) is an overall rare, but highly lethal cancer entity [1,2]. Globally, BTC is much more common in Asia with incidence rates of more than 80 persons per 100,000 per year, e.g., in Thailand compared to an incidence of approximately 1–2 person per 100,000 in Western countries [3]. Recent epidemiological studies have shown a general increase in incidence over the last 20 years [4,5]. The occurrence of BTC is associated with a variety of liver- and biliary tract-damaging diseases such as gall- and bile duct stones and recurrent cholangitis, hepatitis B or C infection, primary sclerosing cholangitis (PSC), parasite infection (especially of Opisthorchis viverrini) as well as all kinds of chronic liver diseases such as alcoholic and non-alcoholic liver diseases [2,6,7]. Risk factors are also not evenly distributed worldwide. In Western countries, BTC is more common in the elderly (>65 years of age) and in men [2,8].

1.1. Systematic of Biliary Tract Cancers

The biliary tract (also called biliary tree or biliary system) is the generic term for the biliary ducts, the gallbladder and the common hepatic duct. Due to the anatomy of the biliary tract, the term “biliary tract cancers” summarizes a heterogeneous group of tumors. Carcinomas of the biliary ducts and the common hepatic duct are named cholangiocarcinoma (CCA) referring to its origin, the cholangiocytes, specialized epithelial cells lining the biliary tract [9]. More than 90% of CCA are mucin-producing, well-differentiated adenocarcinomas [2]. CCA can occur in all parts of the biliary tract, and a distinction is made between intrahepatic CCA and extrahepatic CCA (perihilar or distal; see Figure 1) [3,9,10]. The different types of CCA share molecular patterns that are relevant to systemic therapy, but the distinction between intrahepatic, perihilar and distal CCA is of high relevance as surgical treatment and prognosis differs significantly [11]. Gallbladder carcinomas are usually named separately from other CCA due to their particularly aggressive tumor biology [12]. Furthermore, unlike CCA, gallbladder carcinoma occurs more frequently in women than in men [8,13].

1.2. Diagnosis, Therapy and Prognosis of Biliary Tract Cancers

Early clinical manifestation of all BTCs is unspecific or even asymptomatic [2]. Advanced-stage disease goes along with jaundice, cholestasis and cholangitis depending on the tumor site (Figure 1) [2,14]. Several publications emphasize the necessity to expect BTC in patients with liver and biliary tract diseases, but structured concepts for early detection of CCA and gallbladder carcinoma are widely lacking [9,14,15].
Therapy decision is usually made multidisciplinary and mostly combines hepatobiliary surgery and (adjuvant) systemic chemotherapy in case of resectability [3,16,17,18]. The only potential curative therapeutic approach is the complete surgical tumor resection, but research indicates that curative-intent surgery is only possible in approximately a third of patients [19,20,21]. Because tumor resection requires major hepatobiliary surgery, surgery is associated with a significant risk of complications bearing also the possibility of liver failure or other severe postoperative complications [22,23]. Even though surgery is the only approach to cure CCA and gallbladder carcinoma, long-term survival rates after surgery are still poor, mainly due to rapid disease recurrence [13,17,19]. Recent research indicates that more than one in two CCA patients develop recurrence after curative-intent surgery [24,25,26,27].

1.3. The Gut–Liver Axis

Even more than most of the other organs, the liver is in direct contact with the gut microbiota due to the enterohepatic circulation. The portal vein transports gut-derived metabolites to the liver [28]. In turn, the liver is able to “communicate” with the gut microbiota by secretion of bile acids and other molecules which shape the microbial environment (Figure 2).
In 2022, Binda et al. recapitulated three mechanisms of cancer-inducing gut microbiota activity: first, bacterial toxins and metabolites; second, modulation of the host’s local and systemic immune response; third, metabolic changes in the microbiota and the host [29]. The gut–liver axis has a bidirectional relationship that allows communication between liver cells and bile duct cells and the gut microbiota. Therefore, it can be assumed that the mechanisms described by Binda et al. can also play a role in promotion and progression of liver cancer and BTC. Several publications emphasize the role of gut microbiota in in liver diseases and in carcinogenesis. The development of PSC in patients with ulcerative colitis might be provoked by gut microbiota dysbiosis [30,31,32,33]. Furthermore, it has been suggested that gut microbiota dysbiosis might be also able to promote carcinogenesis [34,35,36]. As early as 2012, it was suggested that Escherichia coli and its metabolites may contribute to inflammation that promotes the development of colorectal cancer [37]. A potent genotoxin of certain strains of Escherichia coli, named colibactin, is able to induce DNA damage in human cells [38]. In 2013, Yoshimoto et al. were able to show that obesity-associated changes in gut microbiota contribute to an increase in intestinal deoxycholic acid, which is known to cause DNA damage. By the enterohepatic circulation, deoxycholic acids provokes a pro-inflammatory type of hepatic stellate cells, which contribute to development of liver cancer in mice [39]. Dapito et al. showed that the toll-like receptor-driven promotion of hepatocellular carcinoma (HCC) in mice is affected by ligands of the intestinal microbiota [40]. Several publications were able to promote data about the role of gut microbiota in HCC genesis, but the evidence for CCA and gallbladder carcinoma is still not clarified. The role of gut microbiota in CCA and gallbladder carcinoma is plausible, but recommendations for considering gut microbiota in BTC patients are still lacking. Therefore, this scoping review aimed to evaluate recent publications regarding the role of the gut microbiota in diagnosis, treatment and prognosis of CCA and gallbladder carcinoma to estimate potential recommendations for gut microbiota handling in BTC patients.

2. Methods

We performed a scoping literature review searching databases of Medline, Cochrane Library and Web of Science from database inception to June 2023. The search terms were “‘biliary cancer’ AND gut microbio*”, “‘biliary carcinoma’ AND gut microbio*”, “‘biliary tract cancer’ AND gut microbio*”, “‘bile duct cancer’ AND gut microbio*”, “cholangiocarcinoma AND gut microbio*”, “‘gallbladder cancer’ AND gut microbio*” and “‘gallbladder carcinoma’ AND gut microbio*”. Only English and German abstracts and manuscripts were evaluable. All types of publications were considered, including human and animal trials, with the exception of conference abstracts, editorials, and methodical publications. We checked the reference lists of all relevant manuscripts for other appropriate publications. If the results of reviews were a repetition of original research already included, the reviews were excluded. To ensure quality of this narrative review, the manuscript was prepared according to the PRISMA Extension for Scoping Reviews and the recommendations of von Elm et al. [41,42].

2.1. Population

All kinds of animal trials as well as adult patients diagnosed with CCA or gallbladder carcinoma, regardless of gender and curative or palliative intention to treat, were eligible for review inclusion. For review evaluation, it was obligatory that publications provided data about the patient composition of gut microbiota, measured by sequencing or conventional stool cultivation. Since the review focused on gut microbiota, data about tissue or bile microbiota were not considered.

2.2. Research Questions

The aim of this scoping review was to evaluate the role of gut microbiota in diagnosis, treatment and prognosis of CCA and gallbladder carcinoma. The following research questions were posed before starting database research:
Do patients with CCA or gallbladder carcinoma show distinct gut microbiota changes compared to healthy controls?
Does the gut microbiota affect the postoperative outcome of CCA or gallbladder carcinoma patients who underwent curative-intent surgery?
Does the gut microbiota affect the chemotherapeutic or systemic treatment response of CCA or gallbladder carcinoma patients?
Does the gut microbiota affect the prognosis and the overall survival of patients suffering from CCA or gallbladder carcinoma?

3. Results

Systematic search yielded 154 results, of which 68 were not suitable for evaluation after screening of abstracts. Further 28 reports were excluded after full-text evaluation. Overall, a total of 12 studies and one systematic review were included. An overview of the search progress is shown in Figure 3. Detailed methods of included trials are shown in a supplementary file (see Supplementary Materials—Methods).

3.1. Do Patients with CCA or Gallbladder Carcinoma Show Distinct Gut Microbiota Changes Compared to Healthy Controls?

In total, data from 282 CCA patients and 268 gallbladder carcinoma patients were evaluated. Two studies used cultivation methods for evaluation of single bacteria [43,44], and seven studies used novel sequencing techniques for evaluation of the entire gut bacteriota (six studies, [45,46,47,48,49,50]) or gut mycobiota (one study, [51]). Most of the studies excluded patients with other gastrointestinal diseases. The timing of measurement varied; four studies reported stool sampling before treatment, one reported mixed timing, and others did not specify the exact timing of measurements. Six studies excluded patients with prior use of antibiotics [46,47,48,49,50,51], one study stated the use of various antibiotics before stool sampling [45] and two studies did not report about prior application of antibiotics [43,44]. An overview of the characteristics of the eight included studies and one systematic review addressing the first research question is shown in Table 1 and Table 2.

3.1.1. Gut Microbiota Changes in CCA Patients

The entire composition of gut microbiota of CCA patients compared to healthy controls was investigated in seven studies [45,46,47,48,49,50,51]. The results of diversity indices were inhomogeneous. One study reported higher α-diversity of CCA patients compared to healthy controls [48]. Two studies stated lower α-diversity of CCA patients compared to healthy controls (one of gut bacteriota [46] and one of gut mycobiota [51]). Three studies found no differences of α-diversity of CCA patients compared to healthy controls [47,49,50], and another study did not measure α-diversity of CCA patients compared to healthy controls [45]. The β-diversities within CCA patients were lower compared to healthy controls in one trial [48], whereas another trial did not report differences of β-diversities within CCA patients compared to healthy controls [45]. Further two trials reported a separated cluster of CCA patients based on β-diversities [46,47].
The composition of gut microbiota differed significantly between CCA patients and healthy controls to various degrees. A simplified overview of differences in gut microbial composition of CCA patients compared to healthy controls is shown in Figure 4.
The differences of gut mycobiota, which were evaluated in one trial by Zhang et al., are reported in Table 2. The authors found a higher abundance of opportunistic pathogenic fungi such as Candida albicans and a lower abundance of potential beneficial fungi such as Saccharomyces cerevisiae in CCA patients compared to healthy controls [51].
Besides comparisons with healthy controls, one study compared gut microbiota composition of CCA patients with patients suffering from primary sclerosing cholangitis (PSC) [45]. The authors reported no differences of diversity indices or taxa abundances between CCA patients and PSC patients with and without CCA.
Two of the publications suggested the use of predictive models to distinguish CCA patients from healthy subjects by evaluating gut microbiota. Zhang et al. suggested that the genera Burkholderia, Caballeronia, Paraburkholderia, Faecalibacterium and Ruminococcus were able to discriminate CCA patients from healthy controls [49]. Deng et al. postulated that the genera Muribaculaceae unclassified, Lachnospiraceae NK4A136 group, Escherichia/Shigella, and Klebsiella might be promising biomarkers for distinguishing CCA patients from patients with hepatocellular carcinoma (HCC) or from healthy controls [47].

3.1.2. Gut Microbiota Changes in Gallbladder Carcinoma

One systematic review and one case–control study were evaluated that address the gut microbial changes in gallbladder carcinoma patients. The studies focused on the impact of Salmonella spp. in the development of gallbladder carcinoma. There were no studies examining the composition of the entire gut microbiota of patients suffering from gallbladder carcinoma.
A systematic review published in 2014 by Nagaraja et al. emphasized the role of Salmonella typhi in development of gallbladder carcinoma [43]. The chronic carrier status as determined by cultivation was associated with gallbladder carcinomas, whereas the past medical history of infection with Salmonella typhi was not associated. A case–control study by Koshiol et al. compared stool samples from patients with gallbladder carcinoma (n = 13), patients with gallstone disease (n = 9) and healthy subjects (n = 13), but found no differences of Salmonella spp. by culture between the groups [44]. The authors added an additional systematic review with meta-analysis evaluating the associations between Salmonella spp. and gallbladder carcinoma, and concluded that the results support a potential role of Salmonella spp. measured by of stool culture in development of gallbladder carcinoma.

3.2. Does the Gut Microbiota Affect the Postoperative Outcome of CCA or Gallbladder Carcinoma Patients Who Underwent Curative-Intent Surgery?

None of the evaluated studies reported about the postoperative outcome of CCA patients or gallbladder carcinoma patients. Therefore, it is not possible to draw any conclusions regarding this research question.

3.3. Does the Gut Microbiota Affect the Chemotherapeutic or Systemic Treatment Response of CCA or Gallbladder Carcinoma Patients?

Two studies with a total of 44 patients evaluated the role of gut microbiota in response to programmed cell death receptor-1 (PD-1) antagonists in patients suffering from CCA [52,53]. Both studies reported the results of patients with advanced-stage disease after failure of first-line treatment. We found no reports evaluating the impact of gut microbiota on chemotherapeutic response of CCA or gallbladder carcinoma patients. An overview of the characteristics of the included studies is presented in Table 3 and Table 4.
The first study by Mao et al. reported on the response of patients with unresectable HCC and advanced biliary tract carcinoma [52]. The authors did not find differences in bacterial diversity indices or abundances between HCC patients and advanced biliary tract carcinoma patients and evaluated the treatment response of both entities together. In the group of patients with a complete or partial response or stable disease of ≤6 months, a higher abundance of phylum Bacteroidota and of order Bacteroidales and a lower abundance of order Veillonellales were found. Furthermore, the relative abundance of Lachnospiraceae bacterium-GAM79 and a few species from the Oscillospiraceae family (Ruminococcus callidus, Gemmiger formicilis, Eubacterium siraeum and Faecalibacterium genus) were significantly enriched in patients with a complete or partial response or stable disease. The authors were able to perform a dynamic fecal sampling (sampling every three weeks before the anti-PD-1 infusion) in eight patients indicating the stability of gut microbiota along with the treatment in patients with a complete or partial response or stable disease.
Jin et al. compared gut microbiota of advanced-stage CCA patients with rapid progress within 6 months to advanced stage CCA patients with a slower progress [53]. The authors reported a higher abundance of phylum Pseudomonadota in patients with a rapid progress. A total of 26 Pseudomonadota species differed significantly between patients with rapid progress and patients with slower progress. Of particular interest were Serratia Marcescens and Raoultella Planticola, and the authors proposed these species as biomarkers of treatment response.

3.4. Does the Gut Microbiota Affect the Prognosis and the Overall Survival of Patients Suffering from CCA or Gallbladder Carcinoma?

Five studies (four human studies and one animal trial) reported on potential prognostic factors of the gut microbiota for promotion or progression of CCA or gallbladder carcinoma. Four studies evaluated CCA patients and one study reported on gallbladder carcinoma patients. The aims of these studies varied as two studies reported on gut microbial alteration in patients with advanced disease compared to patients with limited disease [48,51], and another study evaluated the association between the gut microbiota and the progression-free survival (PFS) and overall survival (OS) of advanced disease patients being treated with programmed cell death receptor-1 [52]. The animal trial focused on the development of CCA in PSC [54], and the last study evaluated epidemiological data regarding the mortality of Salmonella typhi or Salmonella paratyphi carriers [55]. An overview of the included studies is shown in Table 5 and Table 6. Further results of four of the five studies were previously mentioned in other subheadings.
Jia et al. (see also Section 3.1) found higher abundances of family Oscillospiraceae and lower abundances of family Eubacteriaceae as well as of genera Allobaculum, Pediococcus, Pseudoramibacter, and Peptostreptococcus in CCA patients with venous infiltration compared to CCA patients without venous infiltration [48]. Vascular infiltration is known to worsen the outcome of CCA patients [56,57].
Zhang et al. (see also Section 3.1) showed distinct differences of the gut mycobiota composition of patients with advanced-stage CCA (stage III and IV) compared to CCA patients with lower stages (stages I and II) [51]. The authors postulated a potential role of Candida albicans in advanced disease.
Mao et al. (see also Section 3.3) investigated the association between the gut microbiota and the oncological outcome of unresectable HCC and advanced biliary tract carcinoma [52]. Survival analysis revealed longer progression-free survival (PFS) and overall survival (OS) in patients with a higher abundance of species Lachnospiraceae bacterium-GAM79, Erysipelotrichaceae bacterium-GAM147, Ruminococcus callidus, Alistipes megaguti and Bacteroides zoogleoformans. Higher abundance of family Veillonellaceae was negatively associated with the patients’ PFS and OS. The authors separated biliary tract cancer patients from HCC patients and reported that biliary tract cancer patients with a higher abundance of order Bacteroidales had a significantly better PFS and OS. As mentioned before in the overall group analysis, a higher abundance of family Veillonellaceae was also negatively associated with the PFS and OS of biliary tract cancer patients.
Regarding the development of CCA, an animal trial by Zhang et al. emphasized the role of Gram-negative bacteria in CCA progression of patients suffering from PSC [54]. The authors treated mice with bile duct ligation (BDL) and mice with dextran sulfate sodium (DSS)-induced colitis with neomycin to eliminate Gram-negative bacteria, resulting in lower rates of CCA. After fecal transplantation of vancomycin-treated mouse stool in germ-free mice, a higher tumor burden of intrahepatic injected RIL-175 tumor cells (a murine HCC cell line) was found. The results could not be reproduced in mice free of colitis or PSC-like lesions.
The study published by Caygill et al. (see also Section 3.1) almost 30 years ago postulated that carriers of Salmonella typhi or Salmonella paratyphi have a large excess of cancer mortality compared to non-carriers, particularly of gallbladder carcinoma [55].

4. Discussion

In summary, the studies discussed above highlight a possible role of the gut microbiota in diagnosis, therapy and prognosis of CCA and gallbladder carcinoma.

4.1. Limitations of the Included Studies

Regarding the methodology of the studies evaluated, most studies had small sample sizes (on average, 29 CCA patients per study), and proper statistical planning and power analyses were missing. Differences in sequencing techniques and primers made the studies considered hardly comparable. In most of the included studies, only one stool sample was examined, which makes it difficult to draw conclusions due to the dynamic microbial environment, which is affected by the circadian rhythm and other external factors [58].
It is necessary to note that the included publications showed demographic differences of patients. Both studies about the clinical response to programmed cell death receptor-1 treatment provided just limited or no information about further clinical data of patients [52,53]. In four publications, patients suffering from CCA were significantly older and more often male than controls [45,46,49,50]. It is known that age and sex can affect the composition of the gut microbiota [59]. Furthermore, all authors said nothing about the diet of included patients, although food is one of the main modulators of the gut microbiota [60,61].
The majority of studies were conducted in China. Although it can be assumed that the included patients might have similar nutritional and lifestyle habits due to their origin, we found no consistent or overlapping observations between the studies. In general, the results are hardly comparable to those of patients from Western countries as it is known that origin and ethnicity affect the gut microbiota composition [62,63,64].
Most of the studies controlled for application of antibiotics as a major influencing factor of the gut microbial composition. The results of the study by Miyabe et al. are of limited value as different antibiotic therapies have been administered [45]. Due to microbial selection by antibiotics, it is clearly comprehensible that CCA patients with an antibiotic treatment are not comparable to other subjects without an antibiotic treatment.
Mao et al. analyzed data of patients suffering from HCC together with data of CCA patients, which is debatable [52]. Both tumor entities are primary liver cancers, but etiology, treatment modalities, tumor biology, prognosis and at last affected patient demographic data might differ significantly [65,66]. All of the studies refrained from distinguishing between the different sites of biliary tract cancer, although, as mentioned in the introduction, it is known that the location of the tumor has an influence on therapy and prognosis [11].

4.2. Alteration of Gut Microorganisms in CCA

The lack of clear and consistent changes in the microbial diversity indices is interesting, but unsurprising. Despite changes in habits or lifestyle, research indicates high stability of gut microbiota diversity, and an alteration of individual species might not be able to affect the diversity of the entire microbial community [64,67,68]. Noteworthy, the results of our review did not reveal a clear trend towards a lower abundance of health-promoting genera and a higher abundance of potential harmful species in CCA patients. Due to the inconsistent results, it can be hypothesized that the crucial factor for promotion or progression of CCA and gallbladder carcinoma may not be the species, but the metabolites that they provide. The liver is exposed to a variety of bacterial components and metabolites by the enterohepatic circulation, which contribute to the normal function of the human body (Figure 2) [69,70]. However, some of these molecules such as deoxycholic acid can also be harmful [39]. Usually, none of these molecules are exclusively synthesized by one single species. However, some of the possible harmful bacterial metabolites are also useful and contribute to the self-regulating and self-controlling equilibrium of the commensal bacteria [71].
Some of the altered genera observed in this review are discussed in more detail below.

4.2.1. Genera with Lower Abundances in CCA

The included studies reported lower abundances of genera Faecalibacterium, Ruminococcus, Megamonas, Burkholderia, Caballeronia and Paraburkholderia.
The most commonly mentioned species of Faecalibacterium is Faecalibacterium prausnitzii, which is able to produce butyrate and other short-chain fatty acids. It is often considered an indicator for health due to its anti-inflammatory activity [72]. Another potentially harmful species of the same family belonging to the genus Ruminococcus, is Ruminococcus gnavus, which was found to be associated with inflammatory bowel disease and negative cardiovascular health indices [73,74]. Zhai et al. postulated that diarrhea-predominant irritable bowel syndrome might be provoked by Ruminococcus-driven stimulation of the serotonin biosynthesis [75]. However, Ruminococcus is also able to produce butyrate and other short-chain fatty acids [76].
Megamonas appears to be an indicator for the nutritional status of a patient, as studies indicate a higher abundance of Megamonas in obese subjects and a lower abundance in cancer patients suffering from cachexia [77,78]. It remains unclear whether Megamonas might be a sign of a cancer-related cachexia in the patients included in our review. Additionally, Megamonas was found to be negatively correlated with flavonoids, which are discussed to reduce cellular stress [79,80].
Burkholderia encompasses multiple pathogenic species involved in chronic infections [81]. The role of Caballeronia and Paraburkholderia for human health appears to be not clarified at the moment. Liu et al. found higher abundances of Burkholderia, Caballeronia and Paraburkholderia in Uyghur Chinese patients with ulcerative colitis compared to Han Chinese patients with ulcerative colitis [82]. The clinical consequence of this observation remains unclear.

4.2.2. Genera with Higher Abundances in CCA

In the evaluated studies, higher abundances of genera Actinomyces, Alloscardovia, Lactobacillus, Bacteroides, Alistipes, Shigella and Klebsiella were reported in CCA patients.
Species of Actinomyces are known to cause a slowly progressing granulomatous disease named actinomycosis [83]. The infection is endogenous as the natural habitat of Actinomyces is the oral cavity. In addition to actinomycosis, species of Actinomyces can also lead to brain abscesses, or infections of others sites of the body [83]. Although it appears plausible that Actinomyces might be harmful for patients during chemotherapy due to the immunosuppressive situation, there is no evidence for a higher infection risk in chemotherapy-treated patients. Similar to Actinomyces, Alloscardovia omnicolens, a species belonging to the genus Alloscardovia, is sometimes found in infections of poor-conditioned patients [84]. Both genera might be a sign of the poor condition of the included patients.
Interestingly, CCA patients had higher abundances of Lactobacillus, a mutualistic genus that is thought to protect the balance of the human gut microbiota and promote human health [85,86]. One species of Lactobacillus, Lactobacillus plantarum provides indole-3-lactic acid, which ameliorates tumor growth and intestinal inflammation in colorectal cancer mice [87]. Yu et al. reported that Lactobacillus lactis is able to inhibit the non-alcoholic fatty liver disease progression by resorption of its metabolites via the gut–liver axis [88].
The genus Bacteroides is a highly relevant group for health and disease in humans and focus of several research projects. Bacteroides spp. are opportunistic human pathogens causing severe infections and abscesses and being resistant to a variety of antibiotics [89,90]. However, Bacteroides spp. are also mutualistic species protecting the equilibrium of the human gut microbiota from pathogens. Bacteroides spp. are discussed to be able to modulate the immune system, to enhance phagocytosis, to prohibit the colonization of pathogenic species, and to induce the colonization of beneficial species [91]. Animal-based diets and high-fat diets are associated with a higher abundance of Bacteroides spp. [67,92,93]. In mice, Bacteroides spp. is reported to show strong correlations with deoxycholic acid [94]. Bacteroides fragilis belonging to the genus Bacteroides is discussed to be a colorectal cancer-promoting species and a contributor to metastatic disease due to its modulation of cellular adhesion and epithelial tight junctions [95,96,97]. Overall, Bacteroides appears to be a key player, but its role in CCA needs to be further elucidated.
Another interesting and potentially health-promoting genus, which showed a higher abundance in the included CCA patients, is Alistipes. Recent research indicates a lower abundance of Alistipes in liver diseases, accompanied by an increasing reduction in Alistipes in the case of liver disease progression [98]. The higher abundance of this genus in CCA patients is surprising, but the results of Mao et al. underline the impact of Alistipes in these patients as a higher abundance of Alistipes megaguti was associated with a better survival [52]. Nevertheless, Alistipes spp. are associated with mental diseases such as anxiety and eating disorders. It is suggested that Alistipes spp. decreases serotonin by metabolism of the serotonin precursor tryptophan [98,99]. This observation is of interest, as cancer patients have a high risk for developing depression, which is known to affect patient survival [100].
Klebsiella pneumoniae, an ethanol-producing species, is discussed to promote non-alcoholic fatty liver disease due to its metabolites [101]. Species of the genera Klebsiella and Shigella are known to cause severe and potentially life-threatening diseases in humans, but both are also inhabitants of the gut microbiota.

4.3. Clinical Consequences and Future Aspects

Overall, we observed a deficiency in validated and consistent findings across multiple studies and, therefore, we see a lack of high-quality evidence. This lack of consistency in reported bacterial changes, without a clear trend towards either more harmful or more beneficial genera in patients with CCA, along with divergent results observed in different studies, aligns with the methodological inconsistencies. However, gut microbiota research is challenging due to the nature of the gut microbiota. It requires careful research planning that takes into account possible influencing factors and the microbial dynamics. Due to the limitations mentioned above, the analysis of gut microbiota in CCA patients and the usage of predictive models to distinguish between CCA patients and healthy controls is experimental and cannot be recommended at this time. More consistent and evident results might be achieved by larger samples-sizes and by considering influencing factors such as application of antibiotics, age, sex, nutrition, origin and ethnicity in future gut microbiota research. Due to the nature of gut microbiota and its circadian rhythm more longitudinal trials are needed to provide valid results. Experimental studies could investigate the effects of bacterial components and metabolites on the growth of CCA cells. Since our review emphasizes that high-quality data regarding the association between gut microbiota and BTC are widely lacking, both animal and human studies are of future interest.

5. Conclusions

Although evaluated studies show interesting alterations in intestinal microorganisms of patients suffering from BTC, the results are currently still limited by the methodological shortcomings and inconsistent findings mentioned above. Thus, no recommendation can be made at this time to evaluate the gut microbiota in patients with BTC regularly. Future studies need to address these shortcomings in order to clarify whether gut microbiota have an impact on the diagnosis, therapy and prognosis of patients with CCA and gallbladder carcinoma.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/microorganisms11092363/s1. Methods focusing on microbiota analysis of all included studies.

Author Contributions

Writing—original draft preparation and revision: A.-K.L. and H.R.; writing—review and editing: E.K., C.K., M.T.B. and P.K.E.D.; supervision: R.H., F.B. and H.L. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

aCCA: all sites of cholangiocarcinoma or not specified in detail, BTC: biliary tract cancer, BDL: bile duct ligation, CCA: cholangiocarcinoma, DSS: dextran sulfate sodium, HCC: hepatocellular carcinoma, iCCA: intrahepatic cholangiocarcinoma, n. s.: not specified, OS: overall survival, pCCA: perihilar cholangiocarcinoma, PFS: progression-free survival, PSC: primary sclerosing cholangitis, Ref.: reference, spp.: species pluralis.

References

  1. Valle, J.W.; Borbath, I.; Khan, S.A.; Huguet, F.; Gruenberger, T.; Arnold, D. Biliary cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2016, 27, v28–v37. [Google Scholar] [CrossRef]
  2. Squadroni, M.; Tondulli, L.; Gatta, G.; Mosconi, S.; Beretta, G.; Labianca, R. Cholangiocarcinoma. Crit. Rev. Oncol. Hematol. 2017, 116, 11–31. [Google Scholar] [CrossRef] [PubMed]
  3. Rodrigues, P.M.; Olaizola, P.; Paiva, N.A.; Olaizola, I.; Agirre-Lizaso, A.; Landa, A.; Bujanda, L.; Perugorria, M.J.; Banales, J.M. Pathogenesis of Cholangiocarcinoma. Annu. Rev. Pathol. Mech. Dis. 2021, 16, 433–463. [Google Scholar] [CrossRef] [PubMed]
  4. Ellington, T.D.; Momin, B.; Wilson, R.J.; Henley, S.J.; Wu, M.; Ryerson, A.B. Incidence and Mortality of Cancers of the Biliary Tract, Gallbladder, and Liver by Sex, Age, Race/Ethnicity, and Stage at Diagnosis: United States, 2013 to 2017. Cancer Epidemiol. Biomark. Prev. 2021, 30, 1607–1614. [Google Scholar] [CrossRef] [PubMed]
  5. Javle, M.; Lee, S.; Azad, N.S.; Borad, M.J.; Kate Kelley, R.; Sivaraman, S.; Teschemaker, A.; Chopra, I.; Janjan, N.; Parasuraman, S.; et al. Temporal Changes in Cholangiocarcinoma Incidence and Mortality in the United States from 2001 to 2017. Oncologist 2022, 27, 874–883. [Google Scholar] [CrossRef]
  6. Welzel, T.M.; Mellemkjaer, L.; Gloria, G.; Sakoda, L.C.; Hsing, A.W.; Ghormli, L.E.; Olsen, J.H.; McGlynn, K.A. Risk factors for intrahepatic cholangiocarcinoma in a low-risk population: A nationwide case-control study. Int. J. Cancer 2007, 120, 638–641. [Google Scholar] [CrossRef] [PubMed]
  7. Zhang, H.; Yang, T.; Wu, M.; Shen, F. Intrahepatic cholangiocarcinoma: Epidemiology, risk factors, diagnosis and surgical management. Cancer Lett. 2016, 379, 198–205. [Google Scholar] [CrossRef] [PubMed]
  8. Valle, J.W.; Kelley, R.K.; Nervi, B.; Oh, D.-Y.; Zhu, A.X. Biliary tract cancer. Lancet 2021, 397, 428–444. [Google Scholar] [CrossRef]
  9. Razumilava, N.; Gores, G.J. Cholangiocarcinoma. Lancet 2014, 383, 2168–2179. [Google Scholar] [CrossRef]
  10. Khan, S.A.; Tavolari, S.; Brandi, G. Cholangiocarcinoma: Epidemiology and risk factors. Liver Int. 2019, 39, 19–31. [Google Scholar] [CrossRef]
  11. Ahn, D.H.; Bekaii-Saab, T. Biliary cancer: Intrahepatic cholangiocarcinoma vs. extrahepatic cholangiocarcinoma vs. gallbladder cancers: Classification and therapeutic implications. J. Gastrointest. Oncol. 2017, 8, 293–301. [Google Scholar] [CrossRef] [PubMed]
  12. Roa, J.C.; García, P.; Kapoor, V.K.; Maithel, S.K.; Javle, M.; Koshiol, J. Gallbladder cancer. Nat. Rev. Dis. Primers 2022, 8, 69. [Google Scholar] [CrossRef] [PubMed]
  13. Wernberg, J.A.; Lucarelli, D.D. Gallbladder Cancer. Surg. Clin. N. Am. 2014, 94, 343–360. [Google Scholar] [CrossRef] [PubMed]
  14. Hamaoka, M.; Kozaka, K.; Matsui, O.; Komori, T.; Matsubara, T.; Yoneda, N.; Yoshida, K.; Inoue, D.; Kitao, A.; Koda, W.; et al. Early detection of intrahepatic cholangiocarcinoma. Jpn. J. Radiol. 2019, 37, 669–684. [Google Scholar] [CrossRef] [PubMed]
  15. Joo, I.; Lee, J.M. Imaging bile duct tumors: Pathologic concepts, classification, and early tumor detection. Abdom. Imaging 2013, 38, 1334–1350. [Google Scholar] [CrossRef] [PubMed]
  16. Doherty, B.; Nambudiri, V.E.; Palmer, W.C. Update on the Diagnosis and Treatment of Cholangiocarcinoma. Curr. Gastroenterol. Rep. 2017, 19, 2. [Google Scholar] [CrossRef] [PubMed]
  17. Lamarca, A.; Edeline, J.; Goyal, L. How I treat biliary tract cancer. ESMO Open 2022, 7, 100378. [Google Scholar] [CrossRef]
  18. Bitzer, M.; Groß, S.; Albert, J.; Boda-Heggemann, J.; Brunner, T.; Caspari, R.; De Toni, E.; Dombrowski, F.; Evert, M.; Geier, A.; et al. S3-Leitlinie Diagnostik und Therapie biliärer Karzinome. Z. Gastroenterol. 2023, 61, 420–440. [Google Scholar] [CrossRef]
  19. Cillo, U.; Fondevila, C.; Donadon, M.; Gringeri, E.; Mocchegiani, F.; Schlitt, H.J.; Ijzermans, J.N.M.; Vivarelli, M.; Zieniewicz, K.; Olde Damink, S.W.M.; et al. Surgery for cholangiocarcinoma. Liver Int. 2019, 39, 143–155. [Google Scholar] [CrossRef]
  20. Machairas, N.; Lang, H.; Jayant, K.; Raptis, D.A.; Sotiropoulos, G.C. Intrahepatic cholangiocarcinoma: Limitations for resectability, current surgical concepts and future perspectives. Eur. J. Surg. Oncol. 2020, 46, 740–746. [Google Scholar] [CrossRef]
  21. Bartsch, F.; Heinrich, S.; Lang, H. Limits of Surgical Resection for Bile Duct Cancer. Visc. Med. 2015, 31, 189–193. [Google Scholar] [CrossRef] [PubMed]
  22. Mueller, M.; Breuer, E.; Mizuno, T.; Bartsch, F.; Ratti, F.; Benzing, C.; Ammar-Khodja, N.; Sugiura, T.; Takayashiki, T.; Hessheimer, A.; et al. Perihilar Cholangiocarcinoma–Novel Benchmark Values for Surgical and Oncological Outcomes From 24 Expert Centers. Ann. Surg. 2021, 274, 780–788. [Google Scholar] [CrossRef] [PubMed]
  23. Rahbari, N.N.; Garden, O.J.; Padbury, R.; Brooke-Smith, M.; Crawford, M.; Adam, R.; Koch, M.; Makuuchi, M.; Dematteo, R.P.; Christophi, C.; et al. Posthepatectomy liver failure: A definition and grading by the International Study Group of Liver Surgery (ISGLS). Surgery 2011, 149, 713–724. [Google Scholar] [CrossRef] [PubMed]
  24. Koerkamp, B.G.; Wiggers, J.K.; Allen, P.J.; Besselink, M.G.; Blumgart, L.H.; Busch, O.R.C.; Coelen, R.J.; D’Angelica, M.I.; DeMatteo, R.P.; Gouma, D.J.; et al. Recurrence Rate and Pattern of Perihilar Cholangiocarcinoma after Curative Intent Resection. J. Am. Coll. Surg. 2015, 221, 1041–1049. [Google Scholar] [CrossRef] [PubMed]
  25. Komaya, K.; Ebata, T.; Yokoyama, Y.; Igami, T.; Sugawara, G.; Mizuno, T.; Yamaguchi, J.; Nagino, M. Recurrence after curative-intent resection of perihilar cholangiocarcinoma: Analysis of a large cohort with a close postoperative follow-up approach. Surgery 2018, 163, 732–738. [Google Scholar] [CrossRef] [PubMed]
  26. Primrose, J.N.; Fox, R.P.; Palmer, D.H.; Malik, H.Z.; Prasad, R.; Mirza, D.; Anthony, A.; Corrie, P.; Falk, S.; Finch-Jones, M.; et al. Capecitabine compared with observation in resected biliary tract cancer (BILCAP): A randomised, controlled, multicentre, phase 3 study. Lancet Oncol. 2019, 20, 663–673. [Google Scholar] [CrossRef] [PubMed]
  27. Lang, H.; Sotiropoulos, G.C.; Sgourakis, G.; Schmitz, K.J.; Paul, A.; Hilgard, P.; Zöpf, T.; Trarbach, T.; Malagó, M.; Baba, H.A.; et al. Operations for Intrahepatic Cholangiocarcinoma: Single-Institution Experience of 158 Patients. J. Am. Coll. Surg. 2009, 208, 218–228. [Google Scholar] [CrossRef] [PubMed]
  28. Knight, R.; Vrbanac, A.; Taylor, B.C.; Aksenov, A.; Callewaert, C.; Debelius, J.; Gonzalez, A.; Kosciolek, T.; McCall, L.-I.; McDonald, D.; et al. Best practices for analysing microbiomes. Nat. Rev. Microbiol. 2018, 16, 410–422. [Google Scholar] [CrossRef]
  29. Binda, C.; Gibiino, G.; Coluccio, C.; Sbrancia, M.; Dajti, E.; Sinagra, E.; Capurso, G.; Sambri, V.; Cucchetti, A.; Ercolani, G.; et al. Biliary Diseases from the Microbiome Perspective: How Microorganisms Could Change the Approach to Benign and Malignant Diseases. Microorganisms 2022, 10, 312. [Google Scholar] [CrossRef]
  30. Tanaka, A.; Mertens, J.C. Ulcerative Colitis with and without Primary Sclerosing Cholangitis: Two Different Diseases? Inflamm. Intest. Dis. 2016, 1, 9–14. [Google Scholar] [CrossRef]
  31. Bajer, L.; Kverka, M.; Kostovcik, M.; Macinga, P.; Dvorak, J.; Stehlikova, Z.; Brezina, J.; Wohl, P.; Spicak, J.; Drastich, P. Distinct gut microbiota profiles in patients with primary sclerosing cholangitis and ulcerative colitis. World J. Gastroenterol. 2017, 23, 4548. [Google Scholar] [CrossRef] [PubMed]
  32. Nakamoto, N.; Sasaki, N.; Aoki, R.; Miyamoto, K.; Suda, W.; Teratani, T.; Suzuki, T.; Koda, Y.; Chu, P.-S.; Taniki, N.; et al. Gut pathobionts underlie intestinal barrier dysfunction and liver T helper 17 cell immune response in primary sclerosing cholangitis. Nat. Microbiol. 2019, 4, 492–503. [Google Scholar] [CrossRef] [PubMed]
  33. Hov, J.R.; Karlsen, T.H. The microbiota and the gut–liver axis in primary sclerosing cholangitis. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 135–154. [Google Scholar] [CrossRef]
  34. Sepich-Poore, G.D.; Zitvogel, L.; Straussman, R.; Hasty, J.; Wargo, J.A.; Knight, R. The microbiome and human cancer. Science 2021, 371, eabc4552. [Google Scholar] [CrossRef] [PubMed]
  35. Rao, B.; Ren, T.; Wang, X.; Wang, H.; Zou, Y.; Sun, Y.; Liu, S.; Ren, Z.; Yu, Z. Dysbiosis in the Human Microbiome of Cholangiocarcinoma. Front. Physiol. 2021, 12, 715536. [Google Scholar] [CrossRef] [PubMed]
  36. Thomas, A.M.; Manghi, P.; Asnicar, F.; Pasolli, E.; Armanini, F.; Zolfo, M.; Beghini, F.; Manara, S.; Karcher, N.; Pozzi, C.; et al. Metagenomic analysis of colorectal cancer datasets identifies cross-cohort microbial diagnostic signatures and a link with choline degradation. Nat. Med. 2019, 25, 667–678. [Google Scholar] [CrossRef] [PubMed]
  37. Arthur, J.C.; Perez-Chanona, E.; Mühlbauer, M.; Tomkovich, S.; Uronis, J.M.; Fan, T.-J.; Campbell, B.J.; Abujamel, T.; Dogan, B.; Rogers, A.B.; et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 2012, 338, 120–123. [Google Scholar] [CrossRef]
  38. Dziubańska-Kusibab, P.J.; Berger, H.; Battistini, F.; Bouwman, B.A.M.; Iftekhar, A.; Katainen, R.; Cajuso, T.; Crosetto, N.; Orozco, M.; Aaltonen, L.A.; et al. Colibactin DNA-damage signature indicates mutational impact in colorectal cancer. Nat. Med. 2020, 26, 1063–1069. [Google Scholar] [CrossRef]
  39. Yoshimoto, S.; Loo, T.M.; Atarashi, K.; Kanda, H.; Sato, S.; Oyadomari, S.; Iwakura, Y.; Oshima, K.; Morita, H.; Hattori, M.; et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 2013, 499, 97–101. [Google Scholar] [CrossRef]
  40. Dapito, D.H.; Mencin, A.; Gwak, G.-Y.; Pradere, J.-P.; Jang, M.-K.; Mederacke, I.; Caviglia, J.M.; Khiabanian, H.; Adeyemi, A.; Bataller, R.; et al. Promotion of Hepatocellular Carcinoma by the Intestinal Microbiota and TLR4. Cancer Cell 2012, 21, 504–516. [Google Scholar] [CrossRef]
  41. Tricco, A.C.; Lillie, E.; Zarin, W.; O’Brien, K.K.; Colquhoun, H.; Levac, D.; Moher, D.; Peters, M.D.J.; Horsley, T.; Weeks, L.; et al. PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. Ann. Intern. Med. 2018, 169, 467–473. [Google Scholar] [CrossRef] [PubMed]
  42. von Elm, E.; Schreiber, G.; Haupt, C.C. Methodische Anleitung für Scoping Reviews (JBI-Methodologie). Z. Evidenz Fortbild. Qual. Gesundheitswesen 2019, 143, 1–7. [Google Scholar] [CrossRef] [PubMed]
  43. Nagaraja, V.; Eslick, G.D. Systematic review with meta-analysis: The relationship between chronic Salmonella typhi carrier status and gall-bladder cancer. Aliment. Pharmacol. Ther. 2014, 39, 745–750. [Google Scholar] [CrossRef] [PubMed]
  44. Koshiol, J.; Wozniak, A.; Cook, P.; Adaniel, C.; Acevedo, J.; Azócar, L.; Hsing, A.W.; Roa, J.C.; Pasetti, M.F.; Miquel, J.F.; et al. Salmonella enterica serovar Typhi and gallbladder cancer: A case–control study and meta-analysis. Cancer Med. 2016, 5, 3310–3325. [Google Scholar] [CrossRef] [PubMed]
  45. Miyabe, K.; Chandrasekhara, V.; Wongjarupong, N.; Chen, J.; Yang, L.; Johnson, S.; Chia, N.; Walther-Antonio, M.; Yao, J.; Harrington, S.; et al. Potential Role of Inflammation-Promoting Biliary Microbiome in Primary Sclerosing Cholangitis and Cholangiocarcinoma. Cancers 2022, 14, 2120. [Google Scholar] [CrossRef] [PubMed]
  46. Ma, J.; Li, J.; Jin, C.; Yang, J.; Zheng, C.; Chen, K.; Xie, Y.; Yang, Y.; Bo, Z.; Wang, J.; et al. Association of gut microbiome and primary liver cancer: A two-sample Mendelian randomization and case-control study. Liver Int. 2023, 43, 221–233. [Google Scholar] [CrossRef] [PubMed]
  47. Deng, T.; Li, J.; He, B.; Chen, B.; Liu, F.; Chen, Z.; Zheng, J.; Shi, Z.; Zhang, T.; Deng, L.; et al. Gut microbiome alteration as a diagnostic tool and associated with inflammatory response marker in primary liver cancer. Hepatol. Int. 2022, 16, 99–111. [Google Scholar] [CrossRef] [PubMed]
  48. Jia, X.; Lu, S.; Zeng, Z.; Liu, Q.; Dong, Z.; Chen, Y.; Zhu, Z.; Hong, Z.; Zhang, T.; Du, G.; et al. Characterization of Gut Microbiota, Bile Acid Metabolism, and Cytokines in Intrahepatic Cholangiocarcinoma. Hepatology 2020, 71, 893–906. [Google Scholar] [CrossRef]
  49. Zhang, T.; Zhang, S.; Jin, C.; Lin, Z.; Deng, T.; Xie, X.; Deng, L.; Li, X.; Ma, J.; Ding, X.; et al. A Predictive Model Based on the Gut Microbiota Improves the Diagnostic Effect in Patients with Cholangiocarcinoma. Front. Cell. Infect. Microbiol. 2021, 11, 1157. [Google Scholar] [CrossRef]
  50. Ito, Z.; Koido, S.; Kato, K.; Odamaki, T.; Horiuchi, S.; Akasu, T.; Saruta, M.; Hata, T.; Kumagai, Y.; Fujioka, S.; et al. Dysbiosis of the Fecal and Biliary Microbiota in Biliary Tract Cancer. Cancers 2022, 14, 5379. [Google Scholar] [CrossRef]
  51. Zhang, L.; Chen, C.; Chai, D.; Kuang, T.; Deng, W.; Wang, W. Alterations of gut mycobiota profiles in intrahepatic cholangiocarcinoma. Front. Microbiol. 2022, 13, 1090392. [Google Scholar] [CrossRef] [PubMed]
  52. Mao, J.; Wang, D.; Long, J.; Yang, X.; Lin, J.; Song, Y.; Xie, F.; Xun, Z.; Wang, Y.; Wang, Y.; et al. Gut microbiome is associated with the clinical response to anti-PD-1 based immunotherapy in hepatobiliary cancers. J. Immunother. Cancer 2021, 9, e003334. [Google Scholar] [CrossRef] [PubMed]
  53. Jin, S.; Zhao, R.; Zhou, C.; Zhong, Q.; Shi, J.; Su, C.; Li, Q.; Su, X.; Chi, H.; Lu, X.; et al. Feasibility and tolerability of sintilimab plus anlotinib as the second-line therapy for patients with advanced biliary tract cancers: An open-label, single-arm, phase II clinical trial. Int. J. Cancer 2023, 152, 1648–1658. [Google Scholar] [CrossRef]
  54. Zhang, Q.; Ma, C.; Duan, Y.; Heinrich, B.; Rosato, U.; Diggs, L.P.; Ma, L.; Roy, S.; Fu, Q.; Brown, Z.J.; et al. Gut Microbiome Directs Hepatocytes to Recruit MDSCs and Promote Cholangiocarcinoma. Cancer Discov. 2021, 11, 1248–1267. [Google Scholar] [CrossRef] [PubMed]
  55. Caygill, C.P.J.; Hill, M.J.; Braddick, M.; Sharp, J.C.M. Cancer mortality in chronic typhoid and paratyphoid carriers. Lancet 1994, 343, 83–84. [Google Scholar] [CrossRef] [PubMed]
  56. Hanazaki, K.; Kajikawa, S.; Shimozawa, N.; Shimada, K.; Hiraguri, M.; Koide, N.; Adachi, W.; Amano, J. Prognostic factors of intrahepatic cholangiocarcinoma after hepatic resection: Univariate and multivariate analysis. Hepatogastroenterology 2002, 49, 311–316. [Google Scholar] [PubMed]
  57. Bartsch, F.; Baumgart, J.; Hoppe-Lotichius, M.; Schmidtmann, I.; Heinrich, S.; Lang, H. Visceral infiltration of intrahepatic cholangiocarcinoma is most prognostic after curative resection-Retrospective cohort study of 102 consecutive liver resections from a single center. Int. J. Surg. 2018, 55, 193–200. [Google Scholar] [CrossRef]
  58. Voigt, R.M.; Forsyth, C.B.; Green, S.J.; Engen, P.A.; Keshavarzian, A. Circadian Rhythm and the Gut Microbiome. Int. Rev. Neurobiol. 2016, 131, 193–205. [Google Scholar]
  59. Yoon, K.; Kim, N. Roles of Sex Hormones and Gender in the Gut Microbiota. J. Neurogastroenterol. Motil. 2021, 27, 314–325. [Google Scholar] [CrossRef]
  60. Voreades, N.; Kozil, A.; Weir, T.L. Diet and the development of the human intestinal microbiome. Front. Microbiol. 2014, 5, 494. [Google Scholar] [CrossRef]
  61. Zmora, N.; Suez, J.; Elinav, E. You are what you eat: Diet, health and the gut microbiota. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 35–56. [Google Scholar] [CrossRef] [PubMed]
  62. Hasuda, H.; Ikeda, T.; Makizaki, Y.; Yokota, H.; Tanaka, Y.; Ohno, H.; Shimokawa, M.; Matsuoka, H.; Kimura, Y.; Oki, E.; et al. Alterations in the gut microbiome in patients with esophageal carcinoma in response to esophagectomy and neoadjuvant treatment. Surg. Today 2022, 53, 663–674. [Google Scholar] [CrossRef] [PubMed]
  63. Wilson, A.S.; Koller, K.R.; Ramaboli, M.C.; Nesengani, L.T.; Ocvirk, S.; Chen, C.; Flanagan, C.A.; Sapp, F.R.; Merritt, Z.T.; Bhatti, F.; et al. Diet and the Human Gut Microbiome: An International Review. Dig. Dis. Sci. 2020, 65, 723–740. [Google Scholar] [CrossRef] [PubMed]
  64. Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230. [Google Scholar] [CrossRef] [PubMed]
  65. Tischoff, I.; Tannapfel, A. Hepatozelluläres Karzinom und Cholangiokarzinom-Unterschiedliche Prognose, Pathogenese und Therapie. Zentralbl. Chir. 2007, 132, 300–305. [Google Scholar] [CrossRef]
  66. Mejia, J.C.; Pasko, J. Primary Liver Cancers. Surg. Clin. N. Am. 2020, 100, 535–549. [Google Scholar] [CrossRef] [PubMed]
  67. Kohnert, E.; Kreutz, C.; Binder, N.; Hannibal, L.; Gorkiewicz, G.; Müller, A.; Storz, M.A.; Huber, R.; Lederer, A.-K. Changes in Gut Microbiota after a Four-Week Intervention with Vegan vs. Meat-Rich Diets in Healthy Participants: A Randomized Controlled Trial. Microorganisms 2021, 9, 727. [Google Scholar] [CrossRef]
  68. Ding, T.; Schloss, P.D. Dynamics and associations of microbial community types across the human body. Nature 2014, 509, 357–360. [Google Scholar] [CrossRef]
  69. Ohtani, N.; Hara, E. Gut-liver axis-mediated mechanism of liver cancer: A special focus on the role of gut microbiota. Cancer Sci. 2021, 112, 4433–4443. [Google Scholar] [CrossRef]
  70. Chu, H.; Duan, Y.; Yang, L.; Schnabl, B. Small metabolites, possible big changes: A microbiota-centered view of non-alcoholic fatty liver disease. Gut 2019, 68, 359–370. [Google Scholar] [CrossRef]
  71. Lederer, A.-K.; Chikhladze, S.; Kohnert, E.; Huber, R.; Müller, A. Current Insights: The Impact of Gut Microbiota on Postoperative Complications in Visceral Surgery—A Narrative Review. Diagnostics 2021, 11, 2099. [Google Scholar] [CrossRef]
  72. Ferreira-Halder, C.V.; de Sousa Faria, A.V.; Andrade, S.S. Action and function of Faecalibacterium prausnitzii in health and disease. Best Pract. Res. Clin. Gastroenterol. 2017, 31, 643–648. [Google Scholar] [CrossRef] [PubMed]
  73. Hall, A.B.; Yassour, M.; Sauk, J.; Garner, A.; Jiang, X.; Arthur, T.; Lagoudas, G.K.; Vatanen, T.; Fornelos, N.; Wilson, R.; et al. A novel Ruminococcus gnavus clade enriched in inflammatory bowel disease patients. Genome Med. 2017, 9, 103. [Google Scholar] [CrossRef] [PubMed]
  74. Asnicar, F.; Berry, S.E.; Valdes, A.M.; Nguyen, L.H.; Piccinno, G.; Drew, D.A.; Leeming, E.; Gibson, R.; Le Roy, C.; Al Khatib, H.; et al. Microbiome connections with host metabolism and habitual diet from 1,098 deeply phenotyped individuals. Nat. Med. 2021, 27, 321–332. [Google Scholar] [CrossRef] [PubMed]
  75. Zhai, L.; Huang, C.; Ning, Z.; Zhang, Y.; Zhuang, M.; Yang, W.; Wang, X.; Wang, J.; Zhang, L.; Xiao, H.; et al. Ruminococcus gnavus plays a pathogenic role in diarrhea-predominant irritable bowel syndrome by increasing serotonin biosynthesis. Cell Host Microbe 2023, 31, 33–44.e5. [Google Scholar] [CrossRef] [PubMed]
  76. Baxter, N.T.; Schmidt, A.W.; Venkataraman, A.; Kim, K.S.; Waldron, C.; Schmidt, T.M. Dynamics of Human Gut Microbiota and Short-Chain Fatty Acids in Response to Dietary Interventions with Three Fermentable Fibers. MBio 2019, 10. [Google Scholar] [CrossRef] [PubMed]
  77. Ubachs, J.; Ziemons, J.; Soons, Z.; Aarnoutse, R.; Dijk, D.P.J.; Penders, J.; Helvoort, A.; Smidt, M.L.; Kruitwagen, R.F.P.M.; Baade-Corpelijn, L.; et al. Gut microbiota and short-chain fatty acid alterations in cachectic cancer patients. J. Cachexia Sarcopenia Muscle 2021, 12, 2007–2021. [Google Scholar] [CrossRef]
  78. Duan, M.; Wang, Y.; Zhang, Q.; Zou, R.; Guo, M.; Zheng, H. Characteristics of gut microbiota in people with obesity. PLoS ONE 2021, 16, e0255446. [Google Scholar] [CrossRef]
  79. Feng, J.; Zhao, F.; Sun, J.; Lin, B.; Zhao, L.; Liu, Y.; Jin, Y.; Li, S.; Li, A.; Wei, Y. Alterations in the gut microbiota and metabolite profiles of thyroid carcinoma patients. Int. J. Cancer 2019, 144, 2728–2745. [Google Scholar] [CrossRef]
  80. Vissenaekens, H.; Criel, H.; Grootaert, C.; Raes, K.; Smagghe, G.; Van Camp, J. Flavonoids and cellular stress: A complex interplay affecting human health. Crit. Rev. Food Sci. Nutr. 2022, 62, 8535–8566. [Google Scholar] [CrossRef]
  81. Lewis, E.R.G.; Torres, A.G. The art of persistence—The secrets to Burkholderia chronic infections. Pathog. Dis. 2016, 74, ftw070. [Google Scholar] [CrossRef] [PubMed]
  82. Liu, H.; Liu, W.; Huang, X.; Feng, Y.; Lu, J.; Gao, F. Intestinal flora differences between patients with ulcerative colitis of different ethnic groups in China. Medicine 2021, 100, e26932. [Google Scholar] [CrossRef] [PubMed]
  83. Könönen, E.; Wade, W.G. Actinomyces and related organisms in human infections. Clin. Microbiol. Rev. 2015, 28, 419–442. [Google Scholar] [CrossRef] [PubMed]
  84. Takiguchi, Y.; Nagayoshi, M.; Matsuura, Y.; Yokota, S.; Kajiwara, Y.; Akiba, Y. Thoracic empyema caused by Alloscardovia omnicolens and Bifidobacterium dentium. J. Infect. Chemother. 2022, 28, 1567–1570. [Google Scholar] [CrossRef] [PubMed]
  85. Slattery, C.; Cotter, P.D.; O’Toole, P.W. Analysis of Health Benefits Conferred by Lactobacillus Species from Kefir. Nutrients 2019, 11, 1252. [Google Scholar] [CrossRef] [PubMed]
  86. O’Callaghan, J.; O’Toole, P.W. Lactobacillus: Host-microbe relationships. Curr. Top. Microbiol. Immunol. 2013, 358, 119–154. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, Q.; Zhao, Q.; Li, T.; Lu, L.; Wang, F.; Zhang, H.; Liu, Z.; Ma, H.; Zhu, Q.; Wang, J.; et al. Lactobacillus plantarum-derived indole-3-lactic acid ameliorates colorectal tumorigenesis via epigenetic regulation of CD8+ T cell immunity. Cell Metab. 2023, 35, 943–960.e9. [Google Scholar] [CrossRef]
  88. Yu, J.S.; Youn, G.S.; Choi, J.; Kim, C.; Kim, B.Y.; Yang, S.; Lee, J.H.; Park, T.; Kim, B.K.; Kim, Y.B.; et al. Lactobacillus lactis and Pediococcus pentosaceus -driven reprogramming of gut microbiome and metabolome ameliorates the progression of non-alcoholic fatty liver disease. Clin. Transl. Med. 2021, 11, e634. [Google Scholar] [CrossRef]
  89. Wexler, H.M. Bacteroides: The Good, the Bad, and the Nitty-Gritty. Clin. Microbiol. Rev. 2007, 20, 593–621. [Google Scholar] [CrossRef]
  90. Zafar, H.; Saier, M.H. Gut Bacteroides species in health and disease. Gut Microbes 2021, 13, 1848158. [Google Scholar] [CrossRef]
  91. Wang, C.; Zhao, J.; Zhang, H.; Lee, Y.-K.; Zhai, Q.; Chen, W. Roles of intestinal bacteroides in human health and diseases. Crit. Rev. Food Sci. Nutr. 2021, 61, 3518–3536. [Google Scholar] [CrossRef] [PubMed]
  92. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2013, 505, 559–563. [Google Scholar] [CrossRef] [PubMed]
  93. Wan, Y.; Wang, F.; Yuan, J.; Li, J.; Jiang, D.; Zhang, J.; Li, H.; Wang, R.; Tang, J.; Huang, T.; et al. Effects of dietary fat on gut microbiota and faecal metabolites, and their relationship with cardiometabolic risk factors: A 6-month randomised controlled-feeding trial. Gut 2019, 68, 1417–1429. [Google Scholar] [CrossRef] [PubMed]
  94. Zhao, Y.; Wu, J.; Li, J.V.; Zhou, N.-Y.; Tang, H.; Wang, Y. Gut Microbiota Composition Modifies Fecal Metabolic Profiles in Mice. J. Proteome Res. 2013, 12, 2987–2999. [Google Scholar] [CrossRef] [PubMed]
  95. Bartolini, I.; Risaliti, M.; Ringressi, M.N.; Melli, F.; Nannini, G.; Amedei, A.; Muiesan, P.; Taddei, A. Role of gut microbiota-immunity axis in patients undergoing surgery for colorectal cancer: Focus on short and long-term outcomes. World J. Gastroenterol. 2020, 26, 2498–2513. [Google Scholar] [CrossRef] [PubMed]
  96. Mármol, I.; Sánchez-de-Diego, C.; Pradilla Dieste, A.; Cerrada, E.; Rodriguez Yoldi, M. Colorectal Carcinoma: A General Overview and Future Perspectives in Colorectal Cancer. Int. J. Mol. Sci. 2017, 18, 197. [Google Scholar] [CrossRef] [PubMed]
  97. Doocey, C.M.; Finn, K.; Murphy, C.; Guinane, C.M. The impact of the human microbiome in tumorigenesis, cancer progression, and biotherapeutic development. BMC Microbiol. 2022, 22, 53. [Google Scholar] [CrossRef] [PubMed]
  98. Parker, B.J.; Wearsch, P.A.; Veloo, A.C.M.; Rodriguez-Palacios, A. The Genus Alistipes: Gut Bacteria with Emerging Implications to Inflammation, Cancer, and Mental Health. Front. Immunol. 2020, 11, 906. [Google Scholar] [CrossRef]
  99. Prochazkova, P.; Roubalova, R.; Dvorak, J.; Kreisinger, J.; Hill, M.; Tlaskalova-Hogenova, H.; Tomasova, P.; Pelantova, H.; Cermakova, M.; Kuzma, M.; et al. The intestinal microbiota and metabolites in patients with anorexia nervosa. Gut Microbes 2021, 13, 1902771. [Google Scholar] [CrossRef]
  100. Pinquart, M.; Duberstein, P.R. Depression and cancer mortality: A meta-analysis. Psychol. Med. 2010, 40, 1797–1810. [Google Scholar] [CrossRef]
  101. Yuan, J.; Chen, C.; Cui, J.; Lu, J.; Yan, C.; Wei, X.; Zhao, X.; Li, N.; Li, S.; Xue, G.; et al. Fatty Liver Disease Caused by High-Alcohol-Producing Klebsiella pneumoniae. Cell Metab. 2019, 30, 675–688.e7. [Google Scholar] [CrossRef]
Figure 1. Systematics of biliary tract cancers differentiated in intrahepatic, perihilar (also known as Klatskin) and distal cholangiocarcinoma, and gallbladder carcinoma.
Figure 1. Systematics of biliary tract cancers differentiated in intrahepatic, perihilar (also known as Klatskin) and distal cholangiocarcinoma, and gallbladder carcinoma.
Microorganisms 11 02363 g001
Figure 2. Systematics of the gut–liver axis: Bile acids and other molecules secreted by the liver shape the microbial milieu. The gut microbiota delivers cytokines, metabolites and other nutrient molecules via the portal vein.
Figure 2. Systematics of the gut–liver axis: Bile acids and other molecules secreted by the liver shape the microbial milieu. The gut microbiota delivers cytokines, metabolites and other nutrient molecules via the portal vein.
Microorganisms 11 02363 g002
Figure 3. Overview of screening and assessing process. Thirteen reports (twelve trials and one systematic review) were evaluated for review.
Figure 3. Overview of screening and assessing process. Thirteen reports (twelve trials and one systematic review) were evaluated for review.
Microorganisms 11 02363 g003
Figure 4. Simplified overview of gut microbial differences between CCA patients and healthy controls reported by the included trials. Arrows show an increase or decrease in the respective taxon of CCA patients compared to healthy controls. In case of more than one arrow, the taxon was reported in more than one publication.
Figure 4. Simplified overview of gut microbial differences between CCA patients and healthy controls reported by the included trials. Arrows show an increase or decrease in the respective taxon of CCA patients compared to healthy controls. In case of more than one arrow, the taxon was reported in more than one publication.
Microorganisms 11 02363 g004
Table 1. Overview of included original research addressing the first research question “Do patients with cholangiocarcinoma or gallbladder carcinoma show distinct gut microbiota changes compared to healthy controls?”—General information is sorted by year of publication (aCCA = all sites of cholangiocarcinoma or not specified in detail, HCC = hepatocellular carcinoma, iCCA = intrahepatic cholangiocarcinoma, n. s. = not specified, pCCA = perihilar cholangiocarcinoma, PSC = primary sclerosing cholangitis).
Table 1. Overview of included original research addressing the first research question “Do patients with cholangiocarcinoma or gallbladder carcinoma show distinct gut microbiota changes compared to healthy controls?”—General information is sorted by year of publication (aCCA = all sites of cholangiocarcinoma or not specified in detail, HCC = hepatocellular carcinoma, iCCA = intrahepatic cholangiocarcinoma, n. s. = not specified, pCCA = perihilar cholangiocarcinoma, PSC = primary sclerosing cholangitis).
Reference and AuthorYearCountryType of StudyNGroupsExcluded
[43] Nagaraja et al.2014Australiasystematic review255/861Salmonella typhi carrier/non-carriern. s.
[44] Koshiol et al.2016Chilecase–control13/9/13gallbladder cancer/cholelithiasis/healthyn. s.
[48] Jia/Lu/Zeng et al.2019Chinacase–control28/28/16/12iCCA/HCC/cirrhosis/healthymetastatic CCA, mixed-type CCA
[49] Zhang et al.2021Chinacase–control53/47/40aCCA/cholelithiasis/healthyother gastrointestinal or oncological diseases
[47] Deng/Li et al.2022Chinacase–control46/143/40aCCA/HCC/healthyother gastrointestinal or oncological diseases, age > 80 years
[50] Ito et al.2022Japancase–control30/11/10aCCA/BBD */healthycholangitis, severe medical comorbidities, previous treatment +
[46] Ma et al.2023Chinacase–control63/184/40iCCA/HCC/healthyother gastrointestinal or oncological diseases
[45] Miyabe et al.2023USAcase–control11/16/31pCCA #/PSC+CCA/PSCunsuccessful cannulation of bile duct, no bile duct sample, abnormal postsurgical anatomy
[51] Zhang et al.2023Chinacase–control23/17iCCA/healthymixed-type CCA, infectious diseases, other gastrointestinal, autoimmune or oncological diseases
* benign biliary tract disease (cholelithiasis or gallbladder polyps); + including also endoscopic retrograde cholangiography, # not specified in detail, more than 90% pCCA.
Table 2. Overview of included original research addressing the first research question “Do patients with cholangiocarcinoma or gallbladder carcinoma show distinct gut microbiota changes compared to healthy controls?”—Results are sorted by year of publication (see Table 1) (n. s. = not specified, Ref. = Reference).
Table 2. Overview of included original research addressing the first research question “Do patients with cholangiocarcinoma or gallbladder carcinoma show distinct gut microbiota changes compared to healthy controls?”—Results are sorted by year of publication (see Table 1) (n. s. = not specified, Ref. = Reference).
Ref.MethodTime PointAntibiotics or Probioticsα-Diversity *Abundance *
[43]cultivationn. s.n. s.n. s. Salmonella typhi carrier status was associated with gallbladder carcinoma
[44]cultivationn. s.n. s.n. s.no detection of Salmonella spp. (neither in gallbladder cancer patients nor in controls)
[48]16s RNA
(V4)
n. s. recently none↑ family Peptostreptococcaceae
↑ genera Actinomyces, Lactobacillus, Alloscardovia
[49]16s rDNA
(V3-V4)
before treatmentnone for at least 2 monthsno difference↑ family Muribaculaceae
↑ genera Bacteroides, Muribaculum, Alistipes
↓ genera Burkholderia, Caballeronia, Paraburkholderia, Faecalibacterium, Ruminococcus (suggested biomarkers for differentiation between CCA patients and healthy controls)
[47]16s rRNA
(V3-V4)
n. s.none for at least 8 weeksno difference↑ phylum Bacteroidota, family Muribaculaceae
↑ genera Bacteroides, Shigella, Klebsiella, unclassified Lachnospiraceae NK4A136 group
↓ phylum Bacillota, genus Megamonas
[50]16s rRNA
(V3-V4)
before treatmentnone for at least 8 weeksno difference ↑ class Gammaproteobacteria (main family Enterobacteriaceae)
↓ class Clostridia (main family Lachnospiracea)
genera Faecalibacterium and Coprococcus enriched in healthy controls
[46]16s rRNA
(V3-V4)
before treatmentnone for at least 2 months↓ phylum Bacteroidota, family Porphyromonadaceae
[45]16S rRNA
(n. s.)
Mixeddifferent antibioticsn. s. +n. s. +
[51]ITS2 rDNAbefore treatmentnone for at least 3 months↑ phylum Ascomycota, genus Candida (main species Candida albicans), genus Monographella (main species Monographella nivalis)
↓ phylum Mucoromycota, phylum Basidiomycota, genera Saccharomyces (main species Saccharomyces cerevisiae), Pichia (main species Pichia mandshurica), Mucor (main species Mucor circinelloides), Staphylotricum (main species Staphylotricum coccospurum), Actinomucor (main species Actinomucor elegans), Alternaria (main species Alternaria alternata), Fusarium (main species Fusarium oxysporum), Humicola (main species Humicola fuscoatra)
* of cholangiocarcinoma patients compared to healthy subjects; + no comparison to healthy controls, no difference between other groups.
Table 3. Overview of included original research addressing the third research question “Does the gut microbiota affect the chemotherapeutic or systemic treatment response of CCA or gallbladder carcinoma patients?”—General information is sorted by year of publication (BTC = biliary tract cancer, HCC = hepatocellular carcinoma, n. s. = not specified).
Table 3. Overview of included original research addressing the third research question “Does the gut microbiota affect the chemotherapeutic or systemic treatment response of CCA or gallbladder carcinoma patients?”—General information is sorted by year of publication (BTC = biliary tract cancer, HCC = hepatocellular carcinoma, n. s. = not specified).
Reference and AuthorYearCountryType of StudynGroupsExcluded
[52] Mao/Wang/Long/Yang et al.2021Chinacohort 30/35advanced HCC/BTC +n. s.
[53] Jin et al.2023ChinaPhase II
clinical trial
11 *local advanced aCCA
(clinical stage IV)
age < 75 years, cardiac or autoimmune disease,
immunosuppressive treatment
* including 3 patients with a rapid progress and 8 patients with a progression-free survival of more than 6 months. + authors did not differentiate gut microbiota of HCC and aCCA patients.
Table 4. Overview of included original research addressing the third research question “Does the gut microbiota affect the chemotherapeutic or systemic treatment response of CCA or gallbladder carcinoma patients?”—Results are sorted by year of publication (see Table 3) (n. s. = not specified, Ref. = Reference).
Table 4. Overview of included original research addressing the third research question “Does the gut microbiota affect the chemotherapeutic or systemic treatment response of CCA or gallbladder carcinoma patients?”—Results are sorted by year of publication (see Table 3) (n. s. = not specified, Ref. = Reference).
Ref.TreatmentMethodTime PointAntibiotics or Probioticsα-Diversity *Abundance *
[52]anti-PD-1 based systemic therapysequencing, n. s.after failure of first-line therapynone for at least 3 monthsn. s.↑ class Negativicutes, order Enterobacterales, order Veillonellales, family Veillonellaceae
↓ phylum Bacteroidota, order Bacteroidales
[53]sintilimab plus anlotinib16s rRNA
(V3-V4)
after failure of first-line therapyn. s.n. s.↑ phylum Pseudomonadota
suggested species for response:
Serratia Marcescens and Raoultella Planticola
* of patients with rapid progress (<6 months) compared to patients with slower progress.
Table 5. Overview of included original research addressing the fourth research question “Does the gut microbiota affect the prognosis and the overall survival of patients suffering from CCA or gallbladder carcinoma?”—General information is sorted by year of publication (BTC = biliary tract cancer patients, BDL mice = bile duct ligation mice, DSS-colitis mice = dextran sulfate sodium-induced colitis mice, HCC = hepatocellular carcinoma, iCCA = intrahepatic cholangiocarcinoma, n. s. = not specified).
Table 5. Overview of included original research addressing the fourth research question “Does the gut microbiota affect the prognosis and the overall survival of patients suffering from CCA or gallbladder carcinoma?”—General information is sorted by year of publication (BTC = biliary tract cancer patients, BDL mice = bile duct ligation mice, DSS-colitis mice = dextran sulfate sodium-induced colitis mice, HCC = hepatocellular carcinoma, iCCA = intrahepatic cholangiocarcinoma, n. s. = not specified).
Reference & AuthorYearCountryType of StudynGroupsExcluded
[55] Caygill et al. 1994UKcase–control83/386Salmonella carrier/non-carriersubjects with ongoing infection
[48] Jia/Lu/Zeng et al.2019Chinacase–control28/28/16/12iCCA/HCC/cirrhosis/healthymetastatic CCA, mixed-type CCA
[54] Zhang et al. 2021USAexperimental
(including mice)
n. s.BDL mice/DSS-colitis mice/germ-free mice-
[52] Mao/Wang/Long/Yang et al.2021Chinacohort 30/35advanced HCC/BTC +n. s.
[51] Zhang et al.2023Chinacase–control23/17iCCA/healthymixed-type CCA, infectious diseases, other gastrointestinal, autoimmune or oncological diseases
+ authors did not differentiate gut microbiota of HCC and aCCA patients.
Table 6. Overview of included original research addressing the fourth research question “Does the gut microbiota affect the prognosis and the overall survival of patients suffering from CCA or gallbladder carcinoma?”—Results are sorted by year of publication (see Table 5) (PFS = progression-free survival, OS = overall survival, Ref. = Reference, n. s. = not specified).
Table 6. Overview of included original research addressing the fourth research question “Does the gut microbiota affect the prognosis and the overall survival of patients suffering from CCA or gallbladder carcinoma?”—Results are sorted by year of publication (see Table 5) (PFS = progression-free survival, OS = overall survival, Ref. = Reference, n. s. = not specified).
Ref.MethodTime PointAntibiotics or ProbioticsResults
[55]cultivation-n. s.carriers of Salmonella typhi and Salmonella paratyphi had a large excess of cancer mortality, particularly of gallbladder carcinoma (compared to non-carriers)
[48]16s RNA
(V4)
n. s. recently nonein case of venous infiltration ↑ family Oscillospiraceae and ↓ family Eubacteriaceae, genera Allobaculum, Pediococcus, Pseudoramibacter, Peptostreptococcus
[54]16s RNA
(V4)
-nonetreatment with neomycin for elimination of Gram-negative bacteria resulted in fewer CCA, after dysbiotic fecal microbial transplantation germ-free mice developed liver myeloid cell accumulation, which is associated with worse outcome of CCA
[52]sequencing, n. s.n. s.none for at least 3 months↑ species Lachnospiraceae bacterium-GAM79, Erysipelotrichaceae bacterium-GAM147, Ruminococcus callidus, Alistipes megaguti and Bacteroides zoogleoformans
=> longer PFS and OS; ↑ family Veillonellaceae => shorter PFS and OS
In biliary tract cancer patients: ↑ order Bacteroidales => longer PFS and OS, ↑ family Veillonellaceae = shorter PFS and OS
[51]ITS2 rDNAbefore treatmentnone for at least 3 monthsCandida spp. (main species Candida albicans), Dipodascus spp., family Ustilaginaceae, family Clavulinaceae, and Bipolaris spp. CCA patients with stage III–IV compared to those with stage I–II; ↑ class Sordariomycetes, order Xylariales, family Hyponectriaceae, Monographella spp., Annulohypoxylon spp., increased in CCA patients with stage I–II compared to stage III–IV
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lederer, A.-K.; Rasel, H.; Kohnert, E.; Kreutz, C.; Huber, R.; Badr, M.T.; Dellweg, P.K.E.; Bartsch, F.; Lang, H. Gut Microbiota in Diagnosis, Therapy and Prognosis of Cholangiocarcinoma and Gallbladder Carcinoma—A Scoping Review. Microorganisms 2023, 11, 2363. https://doi.org/10.3390/microorganisms11092363

AMA Style

Lederer A-K, Rasel H, Kohnert E, Kreutz C, Huber R, Badr MT, Dellweg PKE, Bartsch F, Lang H. Gut Microbiota in Diagnosis, Therapy and Prognosis of Cholangiocarcinoma and Gallbladder Carcinoma—A Scoping Review. Microorganisms. 2023; 11(9):2363. https://doi.org/10.3390/microorganisms11092363

Chicago/Turabian Style

Lederer, Ann-Kathrin, Hannah Rasel, Eva Kohnert, Clemens Kreutz, Roman Huber, Mohamed Tarek Badr, Patricia K. E. Dellweg, Fabian Bartsch, and Hauke Lang. 2023. "Gut Microbiota in Diagnosis, Therapy and Prognosis of Cholangiocarcinoma and Gallbladder Carcinoma—A Scoping Review" Microorganisms 11, no. 9: 2363. https://doi.org/10.3390/microorganisms11092363

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop