1,965
Views
0
CrossRef citations to date
0
Altmetric
Review

Roles of microbiota in pancreatic cancer development and treatment

, &
Article: 2320280 | Received 06 Nov 2023, Accepted 14 Feb 2024, Published online: 27 Feb 2024

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease with poor prognosis. This is due to the fact that most cases are only diagnosed at an advanced and palliative disease stage, and there is a high incidence of therapy resistance. Despite ongoing efforts, to date, the mechanisms underlying PDAC oncogenesis and its poor responses to treatment are still largely unclear. As the study of the microbiome in cancer progresses, growing evidence suggests that bacteria or fungi might be key players both in PDAC oncogenesis as well as in its resistance to chemo- and immunotherapy, for instance through modulation of the tumor microenvironment and reshaping of the host immune response. Here, we review how the microbiota exerts these effects directly or indirectly via microbial-derived metabolites. Finally, we further discuss the potential of modulating the microbiota composition as a therapy in PDAC.

This article is part of the following collections:
Gut Microbiota in Cancer Development and Treatment

Introduction

Pancreatic ductal adenocarcinoma (PDAC) represents the third leading cause of cancer-related deaths worldwide and is the most common form of pancreatic cancer.Citation1 It is associated with a 5-year survival rate as low as 11% due to its late diagnosis and limited responses to therapy.Citation1,Citation2 Surgical resection followed by adjuvant chemotherapy represents the best treatment approach in PDAC.Citation3 However, most patients present with unresectable or metastatic disease at diagnosis or experience disease relapse after successful surgery.Citation4 Chemotherapy is often administered to locally advancedCitation4–6 or metastaticCitation7–9 patients in combinatorial regimens, including gemcitabine combined with nab-paclitaxel or FOLFIRINOX (5-fluorouracil, leucovorin, oxaliplatin and irinotecan). However, most patients either already exhibit chemotherapy resistance when treatment starts, or develop it over time.Citation10 In addition, novel immune checkpoint blockade therapies that have been revolutionizing the cancer paradigm in several tumor entities do not work in PDAC.Citation11 Likewise, the therapeutic approaches targeting the tumor stroma, which is recognized as one main factor compromising chemotherapy efficacy, have been equally disappointing.Citation12

In the past decades, efforts have been made toward trying to understand how PDAC is initiated and progresses while acquiring such aggressive features. PDAC comprises very heterogeneous tumor phenotypes that differ not only in the tumor cell-intrinsic transcriptional and epigenetic profiles but also in the composition of the stroma and immune cells infiltrating the tumor microenvironment (TME).Citation13 PDAC may originate from either acinar or ductal cells in the exocrine pancreas, giving rise to these different disease subtypes.Citation14–16 In vivo experiments have suggested that Kirsten rat sarcoma virus (KRAS) activation and Tumor protein 53 (TP53) mutations in pancreatic acinar cells activate a process of metaplasia to ductal-like cells, leading to the onset of pancreatic intraepithelial neoplastic (PanIN) lesions that may progress to malignant disease.Citation16,Citation17 However, ductal cells bearing both KRAS and TP53 genetic events may also give rise to the more aggressive basal-like PDAC subtype in an alternative and accelerated process of oncogenesis.Citation15,Citation16 In human disease, it has been reported that more than 90% of PDAC tumors bear KRAS mutations, suggesting that this may represent the initiating event that unleashes malignancy.Citation18 As the disease progresses, tumor cells may collect other genetic mutations, mainly in TP53, mothers against decapentaplegic homolog 4 (SMAD4) and cyclin-dependent kinase inhibitor 2A (CDNK2A) genes.Citation18 However, targeted therapies based on the presence of each one of these mutations have so far been unsuccessful in improving patient care.Citation18

Currently, there is an urgent need to understand which cues dictate treatment efficacy and to improve clinical outcomes of PDAC patients. While the presence of strong homogeneity in key driver mutations in PDAC has long been acknowledged, it is now being recognized that external factors contribute significantly to the heterogeneity of tumor phenotypes observed during disease development. Among these external factors, recent research has put forward the concept that the gut microbiota – a collection of microorganisms residing in the gut – can play a pivotal role in tumor progression and responses to treatment. Indeed, the concept of the microbiota’s central role in maintaining health and homeostasis is not novel, and there has been an increasing association between the microbiota and the development of various diseases throughout the body, extending beyond the gastrointestinal tract.Citation19 Gut dysbiosis has also emerged as a notable phenotypic feature of human PDAC patients over the past decades.Citation20 Sequencing and immunofluorescence studies have contributed toward characterizing the microbiota of PDAC patients, and these associations are being increasingly explored in preclinical models of PDAC. Overall, research suggests that microbes play a role in PDAC carcinogenesis via the promotion of inflammationCitation21–23 and can modulate the response to therapies directlyCitation24 or indirectly via the production of microbiota-derived metabolites.Citation25

In this review, we discuss and summarize what is currently known on how PDAC development and treatment are affected by the microbiota. We provide insights on the current limitations of this field and speculate about promising microbiota-based approaches that may arise to improve the future outcomes of PDAC patients.

Main

The microbiota of PDAC patients differs from healthy controls

The first clues that the microbiota could explain PDAC development arose from observational studies comparing its composition between PDAC patients and healthy controls. By using a combination of 16S rRNA sequencing and shotgun metagenomics, several studies reported that the microbiota signatures of treatment-naïve PDAC patients differ from those of healthy controls across different locations within the human body, id est (i.e.), the oral cavity, the gut and the healthy or diseased pancreas, as reviewed in the following sections.

Composition of the oral microbiota in PDAC patients

Several studies have investigated the oral microbiota of treatment-naïve pancreatic cancer patients compared to healthy controls based on analysis of saliva,Citation26–31 tongue coatCitation32 or oral washCitation33,Citation34 samples. Most studies observed no differences in the alpha diversity of patient oral microbiota compared with controls,Citation27,Citation28,Citation34 except for one where diversity increased in patients with PDAC.Citation30 In general, reports across these cohorts are inconsistent regarding the enrichment of certain bacteria taxa. Despite this, oral samples from PDAC patients were shown to exhibit higher relative abundance of the phylum FirmicutesCitation27,Citation30,Citation32 and Neisseria and Haemophilus genera were consistently less abundant in the saliva of PDAC patients.Citation27–29 In addition to Firmicutes, Fusobacteria and Actinobacteria phyla were also found to be enriched in the tongue coat of pancreatic carcinoma patients,Citation32 although others reported that lower abundance of Fusobacteria in pre-diagnostic mouthwash samples was associated with increased risk of pancreatic cancer.Citation33 Contrasting results have also been observed for the Leptotrichia genus, which belongs to the Fusobacteria phylum and whose abundance was associated with either higherCitation29 or lowerCitation33 risk of pancreatic cancer. Notably, there are few other studies that did not find any differential taxa abundance in the oral cavity when comparing PDAC patients and controlsCitation31,Citation34 or between cases of PDAC and intraductal papillary mucinous neoplasms (IPMN), a potential precursor for pancreatic cancer.Citation27

Although it has been challenging to systematically connect specific pathogens to PDAC across different patient cohorts, the bacterium Porphyromonas gingivalis (P. gingivalis) seems to be closely linked to pancreatic cancer. P. gingivalis is an oral pathogen driving periodontitis and epidemiological studies have indeed suggested a correlation between periodontal disease and pancreatic cancer.Citation35,Citation36 The relative abundance of the Porphyromonas taxa in oral samples of PDAC patients was inconsistently observed to be similarCitation28 or lowerCitation29,Citation32 compared with non-cancer subjects. Even so, Fan et al. identified that the presence of P. gingivalis was associated with increased risk of pancreatic cancer in a prospective study with 361 patients.Citation33 In accordance with this, others have reported that higher levels of circulating antibodies against P. gingivalis in blood samples collected up to 10 years before diagnosis represented a two-fold higher risk of developing pancreatic cancer.Citation37 Besides P. gingivalis, Aggregatibacter actinomycetemcomitans was also linked to a higher risk of developing PDACCitation33 and Farret et al. demonstrated that two other species – Neisseria elongata and Streptococcus mitis – are less present in the saliva of PDAC patients compared to healthy controls.Citation26

Fewer studies have focused on the composition of the fungal community, also referred to as mycobiota, in the oral cavity of PDAC patients. Some of the first observation studies reported that presence of Candida-related oral lesionsCitation38 and confirmed Candida infectionCitation39 correlated with higher PDAC risk in two independent cohorts. More recently, Wei et al. took advantage of Internal Transcribed Spacer (ITS) rRNA sequencing to further characterize the mycobiota in saliva samples from PDAC patients.Citation40 Decreased alpha diversity was found in PDAC patients compared to healthy controls. The taxa Aspergillus and Cladosporium were not only less abundant in PDAC but also presented high classification power to discriminate between both groups.Citation40 However, the study of the mycobiota in PDAC is still in its early stages, and further research is required to solidify this link.

In short, the oral microbiota of PDAC patients is suggested to be distinguishable from healthy patients, but consistent identification and validation of a unique profile of keystone microbes has been difficult. In addition, whether the reported oral dysbiosis precedes or arises after PDAC development remains to be clarified. Notably, it is known that each subregion of the oral cavity hosts a unique microbiotaCitation41 and thus direct comparison between the microbiota signatures of different types of oral samples across PDAC patient cohorts should be avoided. In addition, composition of rare microorganisms varies over time,Citation41 so time of sampling is another factor that may influence these results. Oral microbiota is also highly dependent on lifestyle-related determinants, such as tobacco use,Citation42 which itself represents a risk factor for pancreatic cancer and may thereby act as a confounding factor.Citation1 Considering these challenges, oral-associated microbiota profiles specifically linked to PDAC should be carefully identified. This could be of use in the clinical setting, providing potential biomarkers for noninvasive screening of the disease.

Composition of the intestinal microbiota in PDAC patients

Growing evidence suggests that treatment-naïve PDAC patients also have a unique bacterial signature in the intestinal content. By using a combination of 16S rRNA sequencing and shotgun metagenomics, the fecal microbiota has been characterized in several independent patient cohorts from China,Citation43,Citation44 Japan,Citation30 Israel,Citation45 Spain and Germany.Citation31 Alpha diversity analysis yielded contrasting findings across cohorts, with microbial diversity within PDAC cases being reported to be lower,Citation30,Citation43 higherCitation21 or similarCitation44,Citation45 in comparison with the control group. Despite this, beta-diversity analysis revealed that the composition of the gut microbiota in PDAC was indeed distinguishable from that of healthy subjects.Citation30,Citation31,Citation43–45

When comparing microbial abundance between PDAC patients and healthy controls, different taxa were found to be differentially present in both groups. Overall, lipopolysaccharide-producing bacteria were enriched in the gut of PDAC patients,Citation43 whereas there was a reduction in butyrate-producing bacteria.Citation43,Citation44 Specifically, the Veillonellaceae familyCitation45 and its genus Veillonella ,Citation21,Citation30,Citation31,Citation43,Citation44 as well as the genera Klebsiella ,Citation21,Citation43 Streptococcus Citation30,Citation31,Citation43 and Akkermansia Citation31,Citation45 were consistently identified to be enriched in fecal samples from PDAC patients. In contrast, Bifidobacterium ,Citation31,Citation43 Eubacterium Citation30,Citation44 and Faecalibacterium Citation31,Citation44,Citation45 taxa were more abundant in the gut microbiota of healthy subjects than in PDAC patients. However, contrasting results were reported for other bacteria strains. Prevotella, for instance, was reported to be enriched in PDAC patients compared to healthy subjects in two independent studies,Citation30,Citation43 whereas the opposite was observed in a third cohort.Citation21 Other genera, including Odoribacter ,Citation45 Selenomonas, Hallella, Enterobacter and Cronobacter ,Citation43 were found to be highly abundant in the gut microbiota of PDAC patients in individual studies, but this was not reproduced in the remaining populations.

When comparing the fecal microbiota of PDAC patients with that of patients with pre-malignant lesions, a clear microbial pattern could not be observed between earlier stages and advanced PDAC.Citation45 In a similar trend, the gut microbiota of PDAC was also found to be similar to chronic pancreatitis, a risk factor in pancreatic cancer,Citation30,Citation31 reflecting that microbiota-based early detection of cancer may be challenging. Although most studies carried out so far have not investigated how the intestinal microbiota signature may correlate to responses to treatment, we have recently observed that the pre-treatment microbiota of PDAC patients responding to chemotherapy differs from the microbiota of non-responders.Citation25 In summary, the gut microbiota of PDAC patients is suggested to be distinguishable from healthy patients and to vary according to treatment responses. Hence, it may serve as a diagnostic tool or prognostic marker in the future.

Composition of the intratumoral microbiota in PDAC

The concept of intratumoral microbiota arose from evidence across several cancers suggesting that bacteria could be detected within the tumor microenvironment (TME).Citation46 In the specific case of the pancreas, although it was long considered sterile, a considerable number of studies has now established that bacteria may colonize this organ even under steady state conditions.Citation21–23,Citation31,Citation47

Possible routes for bacteria translocation into the pancreas

Due to the anatomical proximity between the pancreas and intestine, it has been hypothesized that bacteria detected in the pancreas originate in the gastrointestinal tract and translocate via the pancreatic duct. By orally gavaging mice with fluorescently-labeled bacteria, Pushalkar et al. showed that fluorescent bacteria could be detected in the pancreas, possibly through retrograde migration from the duodenum.Citation21 This oral-pancreas migration was further supported by two other studies upon gavage with the green fluorescent protein-labeled fungus S. cerevisiae Citation48 or with the oral pathogen P. gingivalis .Citation49 Indeed, the pancreatic microbiota profile of PDAC patients resembled that of the duodenum, further supporting this hypothesis.Citation47 Proteobacteria, for instance, was shown to be highly abundant in both the duodenum and the pancreas.Citation24,Citation47 In addition, tumors from patients undergoing endoscopic retrograde cholangiopancreatography – a procedure that examines bile duct and pancreatic duct abnormalities, exempli gratia (e.g.), an obstruction – before surgery were shown to have significantly more bacteria in the pancreas than those that were not subjected to this procedure.Citation24 Alternatively, bacteria could also translocate to the pancreas as a result of inflammation-driven increased intestinal permeability.Citation50

However, others have described that no bacteria could be detected in vivo in pancreatic cancer xenografts regardless of antibiotic treatment.Citation22 In addition, bacteria infiltration could not be observed in the pancreas from germ-free mice that had been transferred back to specific pathogen-free (SPF) conditions, nor from interleukin (IL) 10 (IL-10) knockout (KO) mice where the barrier function of the intestinal mucosa is compromised.Citation22 This data suggests that colonization of the pancreas with intestinal bacteria does not happen under physiological conditions, but depends on certain predisposing factors that remain to be identified.

It has also been suggested that microbes may reach the pancreas via other routes, including the lymphatic system. Bacteria may translocate from the intestine to mesenteric lymph nodes independently or via CX3CR1hi mononuclear phagocytes.Citation51,Citation52 From there, they can reach the pancreas as lymphatic drainage takes place.Citation52

Researchers have started to explore how bacteria translocate to the pancreas, but the triggers that unleash this translocation and the mechanisms that enable them to remain there under tumorigenic conditions still need to be elucidated. In addition, the reported antimicrobial properties of the pancreatic fluids and their ability to shape the composition of the intestinal microbiotaCitation53 should be taken into account while investigating this intrapancreatic colonization. Whether bacteria or fungi reach the pancreas as a consequence or as a mediator of carcinogenesis is also largely unknown. The relationship between the microbiota and PDAC is still unclear, and further research is needed to establish causality between the presence of bacteria and tumor progression. Indeed, even though microbiota sequencing has also been employed in prospective clinical studies, the presence of certain bacterial strains could solely act as a marker of cancer-associated inflammatory processes.

Composition of the intratumoral microbiota in PDAC

Using a combination of qPCR and fluorescence in situ hybridization (FISH) against the bacterial 16S rRNA region, Geller et al. first demonstrated that bacterial DNA was present in 76% of a collection of PDAC tumors, as opposed to only 15% of normal pancreas from control donors.Citation24 Supporting these findings, a study conducted by Nejman et al. detected bacterial DNA in 68% of PDAC tumors and showed that the most abundant species in PDAC were members of the Proteobacteria phylum.Citation46 Interestingly, bacterial DNA was mainly detected in the cytoplasm of both cancer cells and immune cells.Citation46 Members of the Gammaproteobacteria were consistently detected in PDAC tissues across studies.Citation24,Citation46,Citation49 Furthermore, high relative abundance of Proteobacteria and Firmicutes phyla in PDAC tumors has also been consistently reported.Citation21,Citation22,Citation46,Citation47,Citation54 Fusobacterium species, which are increasingly being implicated in different gastrointestinal cancers, were detected in 8.8% of the tested PDAC samples and its presence correlated with worse survival rates.Citation55 Fusobacterium nucleatum (F. nucleatum) has also been found in cyst fluid from high-grade IPMN.Citation56 Other studies also compared the microbial abundance in PDAC tumor tissue and adjacent healthy pancreatic tissue. Lactobacillus species (spp.), Akkermansia muciniphila (A. muciniphila) and Bacteroides spp. were found to be enriched in PDAC tumor tissue compared with healthy tissue.Citation31 Despite this, the profiles between both tissues shared some similarities at several taxonomic levels.Citation49

Recent evidence also highlighted a clear difference in the intratumoral microbiota of short-term and long-term survival PDAC patients.Citation54,Citation57 Long-term survival patients from two geographically separated cohorts exhibited a higher microbial alpha-diversity and a unique intratumoral microbiota signature characterized by Saccharopolyspora, Pseudoxanthomonas, Streptomyces and Bacillus clausii .Citation57 Huang et al., however, did not observe differences in alpha-diversity between these groups, but beta-diversity analysis unveiled an enrichment of Sphingomonas, Megasphaera, Bradyrhizobium, Desulfovibrio, Flavobacterium, Enhydrobacter and Megamonas .Citation54 It should be noted that the threshold to define short-term and long-term survivors and the geographic location of each cohort differed between the two studies, which may be the reason for those inconsistencies. Whereas Riquelme et al. considered those surviving more than 5 years after surgery to be long-term survivors,Citation57 the long-term group in the study by Huang et al. included patients with a survival time of longer than 600 days (less than 2 years) after surgery.Citation54

The microbiota of different molecular subtypes of PDAC was also suggested to be different in a study by Guo et al .Citation58 Basal-like (BL) PDAC tumors are poorly differentiated and typically exhibit a more aggressive phenotype accompanied by chemoresistance, whereas the classical subtype is characterized by better therapy responses and overall survival. In this study, the authors carried out FISH against 16S rRNA sequences, demonstrating that BL tumors had an increased bacterial DNA load compared to classical or hybrid subtypes. In addition, Acinetobacter, Pseudomonas and Sphingopyxis genera were highly abundant in the BL subtype and were strongly associated with inflammatory and carcinogenic pathways.Citation58

Fungal DNA was detected in the tumors of PDAC patients in two separate studiesCitation48,Citation59 and was significantly increased in PDAC in comparison with healthy donors.Citation48 The pancreatic mycobiome of patients clustered separately from that of healthy subjects and Malassezia spp. were particularly enriched in the tumor samples.Citation48 By using 18S rRNA sequencing and FISH for fungal DNA, Alam et al further confirmed higher presence of fungal DNA in tumors of KrasG12D Trp53R172H Pdx-Cre (KPC) mice, a mouse model of PDAC, compared with healthy mouse pancreata.Citation60

Other studies reported low amounts of microbial biomass (median of 3682 copies of bacterial DNA/ng of DNA, versus a median of 2766 in the negative control) in pancreatic tissue that were relatively conserved across healthy pancreas, IPMN or PDAC tissues, and therefore unlikely to contribute to malignant transformation.Citation61 Recent reanalysis of previously published microbiotaCitation62 and mycobiotaCitation48 sequencing datasets did not identify the differences reported for PDAC intratumor microbial composition in the original publications.Citation63,Citation64 This highlights the importance of using complementary techniques, such as fluorescent in situ hybridization, in addition to sequencing, to validate the presence of specific bacteria or fungi within PDAC tumors.

Nevertheless, taken together, sequencing data from the different regions throughout the body underlines that patients suffering from PDAC seem to exhibit unique microbial signatures across tissues of the gastrointestinal tract which could serve as diagnostic and prognostic markers. However, there is marked inter-individual variability within each cohort and across different studies, making it more complicated to identify and validate oral, fecal and pancreatic microbiota predictors with potential for PDAC detection. These inconsistencies may be justified by different factors, including: (a) differences in geographic locations and personal lifestyles, which are known to affect the microbiota; (b) small patient sample size and reduced statistical power; (c) challenges in the analysis of microbiota sequencing data from low biomass samples; (d) missing correction for other confounding factors and co-morbidities; (e) differences in sample collection and processing, as well as in methods used for extraction and sequencing of microbial DNA and (f) redundancy of different microbiota taxa. Therefore, larger studies analyzing ethnically and geographically different cohorts of PDAC patients and healthy controls are needed to establish microbiota signatures that might be used as diagnostic markers. Finally, and most importantly, since it is still unclear whether this is the cause or consequence of PDAC development and progression, further mechanistic and functional studies should be performed to evaluate the potential causal role for the microbiota in this disease.

The microbiota promotes pancreatic tumor development in preclinical models

Sequencing studies have contributed greatly to characterizing the unique microbiota signatures of PDAC patients and propose that microbes potentially support PDAC progression. To investigate this putative causative effect, in vivo experiments have been carried out in several animal models of PDAC.

One of the first studies was carried out by Pushalkar et al .Citation21 Here, the authors took advantage of two mouse models: Ptf1aCre LSL-KrasG12D (KC) mice, which spontaneously develop slowly-progressing pancreatic tumors; and the orthotopic model, where tumor cells isolated from the KPC mouse model of PDAC are injected into the pancreas of WT mice. Germ-free KC mice exhibited delayed oncogenesis in the pancreas and, similarly, tumor growth was dramatically reduced in the orthotopic model upon microbial depletion by a cocktail of oral antibiotics composed of vancomycin, neomycin, metronidazole, amphotericin, ampicillin and amphotericin B. Protection from tumor progression in the absence of the microbiota was accompanied by increased tumor infiltration of Th1 CD4+ T cells and cytotoxic CD8+ T cells, reprogramming of tumor-associated macrophages (TAMs) toward higher expression of MHC II, CD86, TNFα, IL-12 and IL-6 and reduced frequencies of myeloid-derived suppressor cells (MDSCs).Citation21 Complementary experiments either depleting CD4+ and CD8+ T cells with neutralizing antibodies or reactivating their function using PD1 antibodies suggested that the beneficial effect of microbiota depletion depends on the induced T cell-mediated immune responses.Citation21

In a similar approach of antibiotic-mediated bacterial ablation in KrasG12DPTENlox/+ mice, Thomas et al. also reported a central role for the gut microbiota in PDAC development.Citation22 Here, the authors used a different antibiotic cocktail administered ad libitum in the drinking water and containing streptomycin, gentamicin, bacitracin and ciprofloxacin. At 3 months of age, antibiotic-treated mice showed lower numbers of malignant lobules than microbiota-intact mice, indicating delayed progression.

In accordance with the data reported by these studies, Sethi et al. observed that microbial depletion by antibiotics also reduced tumor growth in a third model of PDAC consisting of subcutaneous injection of KPC tumor cells.Citation23 Upon ablation of bacteria, there was increased infiltration of interferon-γ (IFN-γ)-producing CD4+ and CD8+ T cells and decreased numbers of IL-17a and IL-10-secreting T cells. Indeed, the protective effect on tumor growth was not present in Rag1 KO mice lacking mature T cells (and B cells) nor after treatment with an IL-17a-neutralizing antibody, underlining that this effect is dependent on a functional adaptive immune system.Citation23

Whereas most studies so far have analyzed the role of the microbiota as a whole in PDAC oncogenesis, others are starting to explore whether this process may be dependent on specific microbes. In fact, considering the classical examples of Fusobacterium nucleatum Citation65,Citation66 and Helicobacter pylori Citation67 being implicated in colorectal and gastric cancer, respectively, it appears plausible that specific bacteria may also have drastic outcomes on tumor development in PDAC. A recent preclinical study solidified the association between P. gingivalis-induced periodontitis and pancreatic cancer observed in human PDAC. P. gingivalis was able to colonize the tumor both in patients and upon oral gavage in an orthotopic model of PDAC.Citation49 Strikingly, orthotopic tumors grew faster in the presence of this bacterium, highlighting for the first time that P. gingivalis can promote PDAC tumorigenesis. This pro-tumor function was mechanistically linked to a CXCR2-mediated infiltration of neutrophils and decreased frequencies of tumor-infiltrating CD8+ T cells and relied on neutrophil-derived elastase.Citation49 In addition, the potential roles of F. nucleatum in PDAC progression have also been addressed.Citation68 F. nucleatum was shown to invade human PDAC cell lines and 3D spheroids, increasing their proliferation and migration capacity. Infection also stimulated secretion of GM-CSF, CXCL1, IL-8 and MIP-3α by both normal and tumor pancreatic cells.Citation68

Within the fungal community, Aykut et al. showed that mycobiome depletion through oral administration of amphotericin B or fluconazole prevented tumor growth in KC mice and in the orthotopic model.Citation48 In addition, Malassezia species were found to be particularly enriched in murine and human PDAC tumors,Citation48 similar to what has been observed in colorectal cancer (CRC).Citation69 Repopulation of antifungal-treated mice with Malassezia globosa but not with other commensal fungi restored carcinogenesis, confirming an active role for Malassezia spp. in PDAC etiology. According to the findings, this effect was dependent on activation of the mannose-binding lectin signaling pathway and of the C3 complement cascade.Citation48 Notably, the antifungal amphotericin B was also included in the experiments performed by Pushalkar et al along with the remaining antibiotics,Citation21 indicating that depletion of both bacteria and fungi might potentially be responsible for the observed effect.

Recently, another study suggested a role of fungi in the pathogenicity of PDAC.Citation60 Alam et al. showed that cancer cell-derived IL-33 triggered recruitment and activation of Th2 and innate lymphoid cells 2 (ILC2) cells to stimulate tumor progression in KPC mice. Importantly, IL-33 secretion, infiltration of Th2 and ILC2 cells and overall tumor burden were highly reduced after amphotericin B treatment. In contrast, repopulation with M. globosa or A. alternata had the opposite effect, suggesting that fungi can promote tumor growth via IL-33-mediated remodeling of the TME.Citation60

Altogether, it is becoming increasingly clear that gut microbes may directly control PDAC onset and progression, but the specific molecular mechanisms underlying these effects are still being elucidated. Considering that bacteria and fungi highly depend on each other,Citation70 it would be relevant to explore whether these pathogenic roles require interactions between both communities. In addition, whether general dysbiosis is the main factor contributing to tumorigenesis or whether specific microbes act as carcinogens in PDAC still needs to be clarified.

Microbiota plays a role in PDAC therapy efficacy and resistance

Besides contributing to pancreatic tumor development, growing evidence suggests that the microbiota may also modulate therapy efficacy and toxicity by affecting drug pharmacokinetics, influencing the host metabolic environment or modulating the composition of the tumor milieu.Citation71,Citation72 Considering that FOLFIRINOX and gemcitabine/nab-paclitaxel are the main therapy regimens applied in PDAC patients, we will focus on these therapies in the following sections.

Influence of the microbiota on chemotherapy efficacy

Taking advantage of the Caenorhabditis elegans (C. elegans) model and the fact that it uses bacteria as a natural food source, it has been shown that distinct bacteria have differential effects on the host response to chemotherapeutic agents.Citation73,Citation74 In fact, specific gut microbes have been described to orchestrate resistance of cancer cells to different chemotherapeutic agents, including oxaliplatin, 5-fluorouracil (5-FU) and gemcitabine. In the context of CRC, for instance, F. nucleatum promotes resistance to oxaliplatin and/or 5-FU regimens, either by activating autophagy in tumor cellsCitation75 or by upregulating BIRC3 expression to prevent apoptosis.Citation76 Retrospective studies in CRC patients also showed that antibiotic treatment improves the efficacy of oxaliplatin but not irinotecan, although the causes behind this specificity are unclear.Citation77

In contrast, others have proposed that the intestinal microbiota mediates treatment efficacy. In a CRC mouse model, the gut microbiota was demonstrated to ensure responses to 5-FUCitation78 or oxaliplatin via induction of anti-tumor immune responses.Citation79 Research conducted by Geller et al. has also demonstrated that Gammaproteobacteria expressing the long isoform of the enzyme cytidine deaminase metabolize gemcitabine to its inactive form, thereby promoting resistance in CRC.Citation24 In a mouse model of CRC, intratumoral presence of Gammaproteobacteria induced resistance to gemcitabine, which was reverted by antibiotic treatment.Citation24

Although the roles of the microbiota in treatment outcomes have not been studied extensively in PDAC yet, it can be speculated that some of the aforementioned CRC-related mechanisms may also apply to PDAC, considering that (a) the same chemotherapy regimens are also used for PDAC treatment; and (b) F. nucleatum and Gammaproteobacteria have also been identified to be present in PDAC patients.Citation24,Citation46 However, Fulop et al. found no association between perichemotherapeutic administration of antibiotics and survival in PDAC patients treated with 5-FU. In contrast, antibiotic treatment correlated with improved survival among patients receiving first-line gemcitabine, suggesting that microbiota-mediated resistance to gemcitabine may also occur in PDAC.Citation80 Similar observations were also reported in an independent retrospective study highlighting that survival rates during gemcitabine-containing regimens were higher when patients were treated with antibiotics.Citation81 Besides bacteria, the fungal community was also reported to orchestrate efficacy of gemcitabine-based chemotherapy, since in vivo depletion of the mycobiome with antifungals was shown to potentiate responses of orthotopic PDAC tumors to chemotherapy.Citation48

Altogether, these findings support that the microbiota can affect the outcomes of patients treated with chemotherapy regimens. However, there is high interindividual variability and redundancy of microbiota taxa and the interplay between the host and the microbiota is remarkably dynamic and complex, involving not only direct microbiota-tumor interactions, but also indirect interactions via the immune system. Due to this, more research is needed to better characterize the mechanisms employed by the microbiota to control chemotherapy responses in PDAC and identify microbiota-based biomarkers that may define better therapy outcomes.

Influence of the microbiota on chemotherapy toxicity

Microbes may also play a role in chemotherapy side effects. Irinotecan is a prodrug that needs to be metabolized by hepatic and intestinal carboxylesterases to form its bioactive metabolite SN-38, responsible for the potent anti-tumor activity.Citation82 SN-38 is subsequently detoxified in the liver by glucuronyltransferases, resulting in the inactive glucuronide SN-38 metabolite (SN-38 G). However, SN-38 G is excreted into the gut via the bile, where bacterial β-glucuronidases can hydrolyze SN-38 G back to the toxic form SN-38. β-glucuronidase-producing bacteria essentially belong to the Bacteroidetes, Firmicutes, Verrucomicrobia and Proteobacteria phyla, which were reported to be enriched in PDAC patients, as previously described.Citation21,Citation22,Citation24,Citation27,Citation30,Citation32,Citation45–47,Citation54 Accumulation of SN-38 in the gut results in delayed severe diarrhea which may even influence continuation of treatment.Citation82 Different strategies have been tested to prevent reactivation of this compound. The use of β-glucuronidase inhibitors, for instance, has been shown to attenuate irinotecan-induced diarrhea without harming the host cells.Citation83,Citation84 Lower intestinal toxicity has also been reported upon use of broad-spectrum antibiotics or under germ-free conditions.Citation85 However, the prophylactic use of antibiotics, for example neomycinCitation86–88 and clarithromycin,Citation89 to prevent diarrhea incidence is controversial.

In addition, the microbiota may also mediate irinotecan toxicity via other β-glucuronidase-independent mechanisms. Streptomycin treatment was shown to be efficient in reducing diarrhea in rats by inhibiting intestinal absorption of irinotecan.Citation90 Importantly, gastrointestinal mucositis and diarrhea are not exclusive to irinotecan and have also been reported upon 5-FU treatment with active participation of the gut microbiota.Citation91

Hyperalgesia, defined by extreme sensitivity to pain, is a common adverse reaction in oxaliplatin-treated patients that was shown to be induced by the gut microbiota.Citation92 It has been shown that germ-free or SPF mice treated with antibiotics did not develop mechanical hyperalgesia in contrast with controls in a mechanism partly dependent on the LPS-TLR4 axis. Conventionalization of GF mice to SPF conditions restored hyperalgesia, further supporting that the gut microbiota contributes to this oxaliplatin-induced effect.Citation92

Chemotherapy-induced dysbiosis

Dysbiosis of the gut microbiota is often observed following chemotherapy and may paradoxically affect the anti-tumor properties of the treatment. In a pancreatic cancer xenograft mouse model, gemcitabine was shown to decrease the proportion of Firmicutes and Bacteroidetes while increasing the relative abundance of Proteobacteria and Verrucomicrobia.Citation93 Other studies have also reported gut dysbiosis after 5-FU treatment. In a CRC model, Yuan et al. described decreased alpha diversity and Actinobacteria abundance in the 5-FU-treated group, whereas no differences were observed in the Firmicutes and Bacteroidetes proportion.Citation78 In contrast, there was decreased Firmicutes and increased Bacteroidetes and Verrucomicrobia abundances in a model of 5-FU-driven intestinal mucositis.Citation94

Although most of these studies focused on the gut microbiota, chemotherapy may also affect the composition of the biliary microbiome. A retrospective study has shown that PDAC patients receiving neoadjuvant therapy (NT) including gemcitabine-based or FOLFIRINOX regimes prior to surgical resection had a distinct bile microbiota compared to primary surgery alone.Citation95,Citation96 However, although marked resistance to cephalosporins was observed in the NT-treated group and in patients undergoing surgery with a biliary stent,Citation95 the opposite was later described by Nadeem et al .Citation97

Overall, the current state of the art suggests that chemotherapy modulates the composition of the microbial communities within the gastrointestinal tract. Considering how the microbiota itself may also determine chemotherapy efficacy, as described earlier, this therapy-induced dysbiosis may paradoxically be a factor that explains their limited therapeutic success and the inevitable chemoresistance observed in PDAC.

Influence of the microbiota on immunotherapy

Immunotherapies, including immune checkpoint blockade with anti-PD-1, anti-PD-L1 and anti-CTLA-4, have been revolutionizing cancer therapies. However, these have not shown promising results in PDAC so far.Citation11 The relationship between the microbiota and the immune system was among the earliest to be investigated. It is now widely acknowledged that the microbiota plays a significant role in modulating immune responses and, as a result, it has been recognized as a potent influencer of the effectiveness of immunotherapies. Efficacy of CTLA-4 blockade with ipilimumab was shown to be compromised under germ-free conditions or after administration of a cocktail of broad-spectrum antibiotics in mouse models of MCA205 sarcoma, melanoma and colon cancer.Citation98 Remarkably, oral administration of Bacteroides species was sufficient to restore the anti-tumor properties of anti-CTLA-4 treatment in an immune-activating process that involved induction of Th1 responses and maturation of dendritic cells within the tumor. It has also been shown that the microbiota modulates the anti-tumor immune responses of PD-1/PD-L1-based immunotherapy in subcutaneous or spontaneous melanoma and MCA205 sarcoma.Citation99,Citation100 Specifically, Bifidobacterium ,Citation99 A. muciniphila and Enterococcus hirae (E. hirae) Citation100 increased the efficacy of anti-PD-1 treatment in vivo. These effects were associated with dendritic cell maturation and IL-12 secretion, enhanced priming and intratumoral infiltration of CD8+ T cells and recruitment of CCR9+CXCR3+CD4+ T cells to the tumor, underlining that the microbiota affects treatment responses via the immune system. A similar effect was also reported for Lactobacillus rhamnosus GG, which improved the responses to anti-PD-1 in melanoma and colon cancer mouse models via CD8+ T cell activation and enhanced IL-6 and IFN-γ production.Citation101 Colonization of germ-free MC38 tumor-bearing mice with a mixture of IFN-γ-producing bacteria further corroborated a better efficacy of anti-PD-1 treatment induced by the microbiota.Citation102 Recently, two studies highlighted a causal role for the microbiota in mediating immunotherapy efficacy in melanoma patients.Citation103,Citation104 Here, transplantation of gut microbiota isolated from responder patients combined with anti-PD-1 treatment induced responses in patients that were previously anti-PD-1-refractory.Citation103,Citation104 Further corroborating the bidirectional interaction between the microbiota and therapies, immunotherapy with anti-CTLA-4Citation98 or anti-PD-1Citation100 were shown to trigger changes in the gut microbiota composition both in vivo and in independent cohorts of cancer patients.

Although all this data suggests that modulation of the gut microbiota may represent a novel immunotherapeutic option in combination with immune checkpoint inhibitors, this area is still understudied in PDAC due to the lack of efficacy of immunotherapies. However, what is known so far also seems to support this notion, since in vivo bacterial ablation reprogrammed the microenvironment of PDAC subcutaneous tumors toward a more pro-tumorigenic phenotype.Citation23 Indeed, as mentioned above, in an orthotopic model of PDAC, tumors did not normally respond to anti-PD-1 treatment but were rendered sensitive to it upon microbiota depletion with oral antibiotics.Citation21 This effect was accompanied by increased activation of CD4+ and CD8+ T cells within the TME.Citation21 Accordingly, activation of CD8+ T cells and lower infiltration of CD4+ Foxp3+ cells were observed in another study following human-into-mice transplantation of microbiota from long-term survival PDAC patients, further supporting that the gut microbiota alters the host immune composition.Citation57 Even so, more studies are needed to confirm these effects and evaluate whether microbiota signatures of PDAC patients may be used to predict clinical responses to future immunotherapy approaches.

Overall, this data reinforces that each individual’s gut microbiota composition should be considered as a factor while addressing the therapeutic benefits of immune checkpoint blockade. In the future, an enhanced comprehension of the microbiota in PDAC patients and potential modifications based on the microbiota composition may contribute to increased responsiveness to immunotherapies.

Microbiota-derived metabolites may affect PDAC development and treatment

Other than translocating to the pancreatic tumor microenvironment to directly interact with PDAC cells and thereby affect tumor development locally, it has been proposed that pancreatic carcinogenesis may also be mediated by microbes indirectly and even from a distance via the host immune system or through microbial-derived metabolites. These remote functions are supported by observations of lower tumor burden upon microbial depletion even in subcutaneous PDAC xenograft models lacking intratumoral bacteria.Citation22 This phenotype was also accompanied by increased infiltration of immune cells in the tumor xenografts even in the absence of local bacteria.Citation22 Thus, it is plausible that these extra-pancreatic effects arise through immune pathways. In fact, the Bifidobacterium-driven anti-tumor functions described by Sivan et al. were also suggested to occur without the need for translocation, since the bacterium was not detected in the mesenteric lymph nodes, spleen or tumors.Citation99

Considering this, researchers started to hypothesize that, besides microbes themselves, microbiota-derived metabolites could also play a role in cancer. To date, the microbiota-derived factors that have been most studied in PDAC lie within five main metabolite groups of metabolites widely known for their immunomodulatory properties,Citation105 comprising short-chain fatty acids (SCFAs), indole derivatives, bile acids (BAs), polyamines and purines. Among SCFAs, in vitro studies have reported anti-proliferative, anti-fibrogenic and pro-apoptotic effects of butyrate (a SCFA) against PDAC cells.Citation106–108 In vivo oral administration of sodium butyrate alone or in combination with gemcitabine to mice bearing subcutaneous PDAC xenografts induced stroma remodeling and was suggested to decrease tumor growth, although without statistical significance.Citation108 In accordance with this, treatment of the colorectal cancer cell line HCT116 with sodium butyrate also inhibited tumor cell growth in vitro and promoted epigenetic methylation reprogramming.Citation109 A recent study demonstrated a synergistic effect of butyrate with oxaliplatin in preventing proliferation, invasion and metastasis of CRC cells in vitro and in vivo .Citation109 A protective role for butyrate and the other two main SCFAs (propionate and acetate) was also reported in 5-FU-induced gastrointestinal mucositis by dampening excessive inflammation.Citation110 In PDAC patients, the relative abundance of butyrate-producing bacteria in the gut was found to be decreasedCitation43,Citation44 and lower concentrations of butyrate in fecal samples of PDAC patients were also reported.Citation44 Overall, this data suggests that the lower levels of butyrate in PDAC patients may contribute to the high tumor proliferation rates and high rates of therapy resistance, although this has to be thoroughly validated in future studies. If this link can be confirmed, using butyrate or butyrate-producing bacteria as an adjuvant therapy to improve chemotherapy efficacy while also controlling its toxicity might be a future application.

Indoles are also increasingly being linked to PDAC. In the gut, metabolism of dietary tryptophan by the microbiota generates indole derivatives, including indole-3-acetic acid (3-IAA) and indole-3-propionic acid (3-IPA). Several studies have explored how the tryptophan metabolism influences the composition of the gut microbiota and intestinal immunity.Citation111 Recently, a pro-tumorigenic role for tryptophan-derived metabolites in PDAC was highlighted by Hezaveh et al .Citation112 In this study, indoles produced by Lactobacillus-mediated catabolism of tryptophan activated the aryl hydrocarbon receptor (AhR) in macrophages. This promoted reprogramming of tumor-associated macrophages into a tumor-supporting phenotype and inhibited IFN-γ expression in CD8+ T cells. Importantly, deleting Ahr in macrophages or removing tryptophan from the diet prevented tumor growth.Citation112 More recently, we have shown that 3-IAA increases the efficacy of FIRINOX (5-FU, irinotecan and oxaliplatin) and gemcitabine+nab-paclitaxel in murine models of PDAC, leading to decreased tumor weight.Citation25 Serum metabolomics unveiled a positive association between 3-IAA and progression-free and overall survival in two human PDAC cohorts. In supplemental murine studies, PDAC tumors were rendered susceptible to chemotherapy upon colonization of germ-free mice with microbiota from responders and oral supplementation of 3-IAA. Mechanistically, this 3-IAA-mediated effect required immune cell-derived myeloperoxidase, which metabolized 3-IAA into toxic products, including 3-methylene-2-oxindole (MOI), that promoted accumulation of reactive oxygen species and autophagy downregulation in the tumor cells, boosting the effect of chemotherapy.Citation25

Besides SCFAs and indole derivatives, BAs have also been explored in the context of PDAC. Primary BAs, including glycocholic acid (GCA), glycochenodeoxycholic acid (GCDCA), taurocholic acid (TCA) and taurochenodeoxycholic acid (TCDCA) are metabolized by gut bacteria upon secretion into the duodenum, giving rise to secondary BAs which include deoxycholic acid (DCA), lithocholic acid (LCA) and ursodeoxycholic acid (UDCA).Citation113 Levels of primary BAs were reported to be higher in PDAC patients,Citation114 especially in those with obstructive jaundice.Citation115 In vitro treatment of Capan-1 and BxPC-3 PDAC cell lines with BAs induced their proliferation, adhesion to collagen 1, migration, invasion and colony-forming ability.Citation115 BAs also upregulated the expression of mucin 4 in a dose-dependent manner and a siRNA-mediated knockdown of mucin 4 prevented these carcinogenic processes.Citation115 Upregulation of cyclooxygenase-2 expression, which is suggested to be linked to cancer development, was also induced in PDAC cell lines upon exposure to DCA and chenodeoxycholic acid.Citation116 In the specific case of DCA, it has also been reported to have oncogenic functions in PDAC via activation of epidermal growth factor receptor (EGFR), Signal transducer and activator of transcription 3 (STAT3) and mitogen-activated protein kinase (MAPK) signaling pathways, promoting cell cycle progression.Citation117 Moreover, the effects of bile itself (which is mainly composed of BAs) in PDAC peritoneal metastasis have also been investigated. Bile samples isolated from PDAC patients reduced peritoneal tumor growth upon co-injection of Panc02 tumor cells and bile into the peritoneum.Citation118 However, since analyses of the roles of specific BAs in PDAC oncogenesis have mainly relied on in vitro studies so far, in vivo functional assessment is still required. Besides carcinogenesis, whether BAs modulate treatment responses in PDAC has not been fully addressed, except for a study by Yang et al., which demonstrated that extracellular TCA decreases the sensitivity of Panc-1 and CFPAC-1 cells to gemcitabine via the S1PR2-ERK pathway.Citation114

The metabolism of polyamines such as putrescine, spermidine and spermine is commonly dysregulated across different tumor entities and has also been implicated in PDAC. Levels of polyamines were found to be enriched in serum of PDAC patients and in KPC mice during disease progression from early stages to PDAC.Citation119 Moreover, expression of polyamine-related genes was inversely associated with survival in a PDAC patient cohort,Citation120 further suggesting a role for polyamines in pancreatic cancer. Indeed, a recent study further explored how PDAC cells synthesize polyamines to ensure tumor growth.Citation121 The authors revealed that glutamine was used in tumor cells as a substrate for ornithine production, which subsequently contributed to polyamine synthesis in a process dependent on ornithine aminotransferase (OAT). Notably, PDAC cells also relied on mutated KRAS to upregulate the expression of proteins required for polyamine synthesis, including OAT.Citation121 Pharmacological targeting of OAT, thus compromising polyamine synthesis, suppressed PDAC growth both in vitro and in vivo.Citation121 Although this study did not specifically investigate bacteria as a source of polyamines, these compounds may also be produced by the gut microbiota.Citation122 Mendez et al., for instance, reported that Lactobacillus reuteri, which was detected in later stages of PDAC development, was associated with polyamine metabolism.Citation119 Overall, this data opens the possibility to target polyamine metabolism in PDAC. However, more research is needed to elucidate to what extent the microbiota contributes to polyamine availability within the TME.

Finally, since purine synthesis is essential for cell proliferation, a dysregulated purine metabolism is often linked to cancer progression.Citation123 Indeed, in PDAC cells, oncogenic KRAS is able to enhance the synthesis of purines and pyrimidines to support proliferation.Citation124 In particular, inosine, xanthine and hypoxanthine, which are degradation products of adenine nucleotides during purine synthesis, were found to be decreased in gemcitabine-treated PDAC-xenografted mice.Citation93 Importantly, the microbiota may also produce these purine-related metabolitesCitation125 and microbial-derived inosine was shown to promote Th1 activation and optimize responses to anti-CTLA-4 treatment in intestinal, bladder cancer and melanoma mouse models.Citation126 However, further research is needed to provide mechanistic insights into the potential roles of microbial-derived nucleosides in PDAC.

Altogether, the biological activity of microbiota-derived metabolites in PDAC still remains largely unknown. However, this is a growing field of interest, and high-throughput metabolomic screens are currently being used to help identify metabolites participating in PDAC development and determining responses to chemo- and immunotherapy.

Microbiota modulation has therapeutic potential in PDAC

Currently, treatment of advanced PDAC patients relies mainly on chemotherapy-based regimens, although resistance to chemotherapy is often observed in the clinics.Citation127 Therefore, PDAC treatment remains a challenge, and there is an urgent need to sensitize PDAC tumors to available therapies or develop novel strategies with better efficacy.Citation128 Considering this scenario and the described roles for microbes in PDAC onset, progression and resolution, modulation of the microbiota may represent a novel therapeutic target to optimize the clinical outcomes in PDAC. So far, there are several approaches to modulate the microbiota which rely on diet, administration of pro- or antibiotics, fecal microbial transplantation, use of bacteriophages and substitution of specific bacteria isolates, among others.

Diet is a key factor influencing the composition of the microbiota in the gut and the availability of bioactive microbial-derived metabolites,Citation129 making dietary interventions an appealing option in microbiota-associated diseases. The association between diet and pancreatic cancer risk is, however, still poorly explored. High-fat diets negatively imbalance the gut microbiota while favoring pathogenic taxa, impairing gut barrier integrity and even driving colorectal carcinogenesis in a microbiota-dependent manner.Citation130 High-fiber diets, in contrast, seem to have anti-tumor properties in CRC as a result of the metabolism of fibers into SCFAs such as butyrate.Citation131 Accordingly, high intake of dietary fibers correlates negatively with pancreatic cancer risk.Citation132 In line with this, in a xenograft mouse model of PDAC, an engineered resistant-starch (ERS) diet, which is fermented into SCFAs by colon bacteria, was shown to delay tumor growth.Citation133 These types of high-fiber non-digestible foods that promote the growth and functions of probiotic bacteria in the gut are often referred to as prebiotics.Citation129

The diet could also be considered an interesting option to improve treatment outcomes in PDAC. Caloric restriction through fasting was described to improve responses to gemcitabine both in vitro and in vivo .Citation134 In addition, we have recently shown that efficacy of chemotherapy in vivo was enhanced by 3-IAA resulting from a short-term tryptophan-high diet, as opposed to a more prolonged administration of dietary tryptophan which promotes tumor growth.Citation25 It seems that dietary changes may even impact therapy toxicity, since dietary fibers were shown to reduce irinotecan-induced toxicity in a rat model of colon carcinoma.Citation135 Even so, despite the promising effects of diet-based interventions in PDAC treatment, we are still lacking solid results from human-based trials. However, dietary clinical trials and clinical implementation of nutritional interventions involve several challenges that should be carefully considered, including difficulties in ensuring patient compliance, absence of standardized protocols and timing and duration of the interventions.Citation136

Probiotics, defined as living microbes associated with a health benefit to the host, are also being considered to ameliorate clinical outcomes in cancer. Research on probiotics in PDAC is still in its early stages, and the available data is scarce. Nevertheless, the most attractive candidates at the moment are butyrate-producing bacteria, such as Faecalibacterium prausnitzii (F. prausnitzii), Eubacterium rectale (E. rectale) and Roseburia intestinalis, given the reported anti-tumor roles of butyrate.Citation137 Interestingly, these three species were shown to be less present in PDAC patients,Citation44 as further confirmed in another study for F. prausnitzii and E. rectale.Citation30 It has also been suggested that the administration of a probiotic mixture (composed mainly of Bifidobacterium spp. and Lactobacterium spp.) to PDAC-xenografted mice may retard epithelial-to-mesenchymal transition in the pancreas.Citation138 In addition to probiotics, the use of postbiotics, consisting of inanimate microorganisms and/or bioactive compounds secreted by the intestinal microbiota (such as SCFAs), may also represent an interesting therapeutic strategy in PDAC, as discussed earlier in section 4. However, research on postbiotics in PDAC is still evolving and their clinical use still faces several challenges, including the limited understanding of their mechanisms of action, as well as the need for standardization within the clinical setting.Citation139

In the case of antibiotics, antibiotic therapy is often used in cancer patients prior to surgery or during treatment as a measure against opportunistic infections.Citation140 The beneficial roles of antibiotics and antifungals in PDAC have been thoroughly analyzed in preclinical modelsCitation21–23,Citation48 and retrospective cohort studies.Citation77,Citation80,Citation141 However, broad spectrum antibiotics may not only eliminate pathogenic bacteria which have been shown to mediate carcinogenesis or resistance to chemo- or immunotherapeutic drugs, but also commensal bacteria that may suppress tumor progression. Therefore, its use as an anti-tumor strategy should be carefully considered. Considering this, bacteriophage therapy is emerging as an alternative to precisely target specific bacteria associated with progression and poor prognosis in cancer,Citation30,Citation142,Citation143 but it has not been formally explored in PDAC yet.

Another possible strategy to modulate the gut microbiota is fecal microbial transplantation (FMT), in which a mixture of gut microbes isolated from a healthy donor is used to colonize the gut of a recipient. Evidence supporting this approach in PDAC has been provided by Riquelme et al., who showed that FMT from patients with long-term survival into mice elicited anti-tumor immune activation characterized by higher numbers of activated CD8+ T cells and reduced infiltration of CD4+Foxp3+ cells and MDSCs.Citation57 To evaluate whether these findings translate into the clinical setting, a phase I trial is ongoing with resectable PDAC patients receiving FMT delivered through colonoscopy or oral capsules, estimated to end in December 2023.Citation144

Concluding remarks and future perspectives

In conclusion, research over the last decade has clearly shown that PDAC is associated with unique oral, gut and pancreatic microbiota signatures (). However, these signatures were not consistent across different cohorts and high inter-individual variability within each cohort was often reported. This poor reproducibility may be due to differential environmental and lifestyle-related factors, including geographical location, smoking and dietary habits, as well as lack of adjustment for potential existing confounding factors and differences in sampling and sequencing methods used. Recently, increasing concerns in the scientific communityCitation63,Citation64 have also reinforced the importance of appropriate and careful sampling and analysis of microbial sequencing datasets, which should include control for sample contamination and correct separation of bacteria or fungi and human genomic reads to minimize false positive findings. Accordingly, despite the vast availability of literature on this topic, identifying a clearly consistent and transversal gut and intratumoral microbial composition still remains a challenge in PDAC. In addition, although experiments conducted in preclinical models of PDAC have been defining several organisms as causative agents of PDAC, more functional experiments are still needed to prove the role of microbe-mediated pathogenicity and thus provide mechanistic evidence of this interplay, especially in the case of fungi.

Figure 1. Roles of the microbiota in PDAC. The microbiota plays a role in PDAC carcinogenesis via the modulation of the immune compartment composition in the tumor microenvironment or via the production of microbiota-derived metabolites. PDAC patients have unique oral, intestinal and intratumoral microbiota signatures and its characterization may be useful as clinical biomarkers of PDAC diagnosis and prognosis. Considering this, strategies modulating the composition of the microbiota have therapeutic potential in PDAC. MDSCs: myeloid-derived suppressor cells. TAMs: tumor-associated macrophages. SCFAs: short-chain fatty acids. BAs: bile acids. Created with BioRender.com.

Figure 1. Roles of the microbiota in PDAC. The microbiota plays a role in PDAC carcinogenesis via the modulation of the immune compartment composition in the tumor microenvironment or via the production of microbiota-derived metabolites. PDAC patients have unique oral, intestinal and intratumoral microbiota signatures and its characterization may be useful as clinical biomarkers of PDAC diagnosis and prognosis. Considering this, strategies modulating the composition of the microbiota have therapeutic potential in PDAC. MDSCs: myeloid-derived suppressor cells. TAMs: tumor-associated macrophages. SCFAs: short-chain fatty acids. BAs: bile acids. Created with BioRender.com.

In addition to the direct roles of microbes themselves, microbial-derived metabolites are also increasingly being explored in this field. In the future, these metabolites may not only become useful as diagnostic biomarkers but also as mediators of therapy efficacy. Importantly, the profile of microbiota-derived metabolites arises from the diverse activities of various bacterial strains, potentially serving as a comprehensive representation of complex bacterial communities. Consequently, investigating these metabolites could aid in mitigating the substantial variability observed when considering the composition of bacteria across different cohorts. Overall, in this review, we have highlighted and summarized the microbial-related functions reported thus far in PDAC () and discussed how these may pave the way for the development of novel diagnostic therapeutic options aiming at improving PDAC prognosis.

Author contributions

MSC wrote the manuscript and prepared the figure. JT and NG co-supervised the work, reviewed and edited the manuscript and the figure.

Acknowledgments

We thank Madeleine Hamley for proofreading the review. This work is supported by a grant from the German Cancer Aid to J.T. and N.G. (70114815). During the preparation of this work the authors used chat.openai.com in order to proofread few sentences. After using this tool, the authors reviewed and edited the content as needed and take full responsibility for the content of the publication.

Disclosure statement

NG reports financial support from F. Hoffmann-La Roche. This is outside the submitted work. All other authors declare no conflict of interest.

Additional information

Funding

The work was supported by the Deutsche Krebshilfe [70114815].

References

  • Kleeff J, Korc M, Apte M, La Vecchia C, Johnson CD, Biankin AV, Neale RE, Tempero M, Tuveson DA, Hruban RH. et al. Pancreatic cancer. Nat Rev Dis Primers. 2016;2(1):1–22. doi:10.1038/nrdp.2016.22.
  • Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin. 2023;73(1):17–48. doi:10.3322/caac.21763.
  • Conroy T, Hammel P, Hebbar M, Ben Abdelghani M, Wei AC, Raoul J-L, Choné L, Francois E, Artru P, Biagi JJ. et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer. N Engl J Med. 2018;379(25):2395–2406. doi:10.1056/NEJMoa1809775.
  • Ducreux M, Cuhna AS, Caramella C, Hollebecque A, Burtin P, Goéré D, Seufferlein T, Haustermans K, Van Laethem JL, Conroy T. et al. Cancer of the pancreas: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26:v56–v68. doi:10.1093/annonc/mdv295.
  • Crane CH, Varadhachary GR, Yordy JS, Staerkel GA, Javle MM, Safran H, Haque W, Hobbs BD, Krishnan S, Fleming JB. et al. Phase II trial of cetuximab, gemcitabine, and oxaliplatin followed by chemoradiation with cetuximab for locally advanced (T4) pancreatic adenocarcinoma: correlation of Smad4(Dpc4) immunostaining with pattern of disease progression. J Clin Oncol. 2011;29(22):3037–3043. doi:10.1200/JCO.2010.33.8038.
  • Mukherjee S, Hurt CN, Bridgewater J, Falk S, Cummins S, Wasan H, Crosby T, Jephcott C, Roy R, Radhakrishna G. et al. Gemcitabine-based or capecitabine-based chemoradiotherapy for locally advanced pancreatic cancer (SCALOP): a multicentre, randomised, phase 2 trial. Lancet Oncol. 2013;14(4):317–326. doi:10.1016/S1470-2045(13)70021-4.
  • Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, Adenis A, Raoul J-L, Gourgou-Bourgade S, de la Fouchardière C. et al. FOLFIRINOX versus Gemcitabine for Metastatic Pancreatic Cancer. N Engl J Med. 2011;364(19):1817–1825. doi:10.1056/NEJMoa1011923.
  • Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, Seay T, Tjulandin SA, Ma WW, Saleh MN. et al. Increased survival in pancreatic cancer with nab-paclitaxel plus Gemcitabine. N Engl J Med. 2013;369(18):1691–1703. doi:10.1056/NEJMoa1304369.
  • Sohal DPS, Kennedy EB, Khorana A, Copur MS, Crane CH, Garrido-Laguna I, Krishnamurthi S, Moravek C, O’Reilly EM, Philip PA. et al. Metastatic pancreatic cancer: ASCO clinical practice guideline update. J Clin Oncol. 2018;36(24):2545–2556. doi:10.1200/JCO.2018.78.9636.
  • Zeng S. Chemoresistance in pancreatic cancer. Int J Mol Sci. 2019;20(18):4504–4519. doi:10.3390/ijms20184504.
  • O’Reilly EM, Oh D-Y, Dhani N, Renouf DJ, Lee MA, Sun W, Fisher G, Hezel A, Chang S-C, Vlahovic G. et al. Durvalumab with or without tremelimumab for patients with metastatic pancreatic ductal adenocarcinoma: a phase 2 randomized clinical trial. JAMA Oncol. 2019;5(10):1431–1438. doi:10.1001/jamaoncol.2019.1588.
  • Ho WJ, Jaffee EM, Zheng L. The tumour microenvironment in pancreatic cancer — clinical challenges and opportunities. Nat Rev Clin Oncol. 2020;17(9):527–540. doi:10.1038/s41571-020-0363-5.
  • Espinet E, Klein L, Puré E, Singh SK. Mechanisms of PDAC subtype heterogeneity and therapy response. Trends Cancer. 2022;8(12):1060–1071. doi:10.1016/j.trecan.2022.08.005.
  • Ferreira RMM, Sancho R, Messal HA, Nye E, Spencer-Dene B, Stone RK, Stamp G, Rosewell I, Quaglia A, Behrens A. et al. Duct- and acinar-derived pancreatic ductal adenocarcinomas show distinct tumor progression and marker expression. Cell Rep. 2017;21(4):966–978. doi:10.1016/j.celrep.2017.09.093.
  • Lee AYL, Dubois CL, Sarai K, Zarei S, Schaeffer DF, Sander M, Kopp JL. Cell of origin affects tumour development and phenotype in pancreatic ductal adenocarcinoma. Gut. 2019;68(3):487–498. doi:10.1136/gutjnl-2017-314426.
  • Flowers BM, Xu H, Mulligan AS, Hanson KJ, Seoane JA, Vogel H, Curtis C, Wood LD, Attardi LD. Cell of origin influences pancreatic cancer subtype. Cancer Discov. 2021;11(3):660–677. doi:10.1158/2159-8290.CD-20-0633.
  • Habbe N, Shi G, Meguid RA, Fendrich V, Esni F, Chen H, Feldmann G, Stoffers DA, Konieczny SF, Leach SD. et al. Spontaneous induction of murine pancreatic intraepithelial neoplasia (mPanIN) by acinar cell targeting of oncogenic Kras in adult mice. Proc Natl Acad Sci U S A. 2008;105(48):18913–18918. doi:10.1073/pnas.0810097105.
  • Hu HF, Ye Z, Qin Y, Xu X-W, Yu X-J, Zhuo Q-F, Ji S-R. Mutations in key driver genes of pancreatic cancer: molecularly targeted therapies and other clinical implications. Acta Pharmacol Sin. 2021;42(11):1725–1741. doi:10.1038/s41401-020-00584-2.
  • Afzaal M, Saeed F, Shah YA, Hussain M, Rabail R, Socol CT, Hassoun A, Pateiro M, Lorenzo JM, Rusu AV. et al. Human gut microbiota in health and disease: unveiling the relationship. Front Microbiol. 2022;13:1–14. doi:10.3389/fmicb.2022.999001.
  • Zhang T, Gao G, Sakandar HA, Kwok LY, Sun Z. Gut dysbiosis in pancreatic diseases: a causative factor and a novel therapeutic target. Front Nutr. 2022;9:1–20. doi:10.3389/fnut.2022.814269.
  • Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, Mohan N, Aykut B, Usyk M, Torres LE. et al. The pancreatic cancer microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 2018;8(4):403–416. doi:10.1158/2159-8290.CD-17-1134.
  • Thomas RM, Gharaibeh RZ, Gauthier J, Beveridge M, Pope JL, Guijarro MV, Yu Q, He Z, Ohland C, Newsome R. et al. Intestinal microbiota enhances pancreatic carcinogenesis in preclinical models. Carcinogenesis. 2018;39(8):1068–1078. doi:10.1093/carcin/bgy073.
  • Sethi V, Kurtom S, Tarique M, Lavania S, Malchiodi Z, Hellmund L, Zhang L, Sharma U, Giri B, Garg B. et al. Gut microbiota promotes tumor growth in mice by modulating immune response. Gastroenterology. 2018;155(1):33–37.e6. doi:10.1053/j.gastro.2018.04.001.
  • Geller LT, Barzily-Rokni M, Danino T, Jonas OH, Shental N, Nejman D, Gavert N, Zwang Y, Cooper ZA, Shee K. et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;1160(6356):1156–1160. doi:10.1126/science.aah5043.
  • Tintelnot J, Xu Y, Lesker TR, Schönlein M, Konczalla L, Giannou AD, Pelczar P, Kylies D, Puelles VG, Bielecka AA. et al. Microbiota-derived 3-IAA influences chemotherapy efficacy in pancreatic cancer. Nature. 2023;615(7950):168–174. doi:10.1038/s41586-023-05728-y.
  • Farrell JJ, Zhang L, Zhou H, Chia D, Elashoff D, Akin D, Paster BJ, Joshipura K, Wong DTW. Variations of oral microbiota are associated with pancreatic diseases including pancreatic cancer. Gut. 2012;61(4):582–588. doi:10.1136/gutjnl-2011-300784.
  • Olson SH, Satagopan J, Xu Y, Ling L, Leong S, Orlow I, Saldia A, Li P, Nunes P, Madonia V. et al. The oral microbiota in patients with pancreatic cancer, patients with IPMNs, and controls: a pilot study. Cancer Causes Control. 2017;28(9):959–969. doi:10.1007/s10552-017-0933-8.
  • Vogtmann E, Han Y, Caporaso JG, Bokulich N, Mohamadkhani A, Moayyedkazemi A, Hua X, Kamangar F, Wan Y, Suman S. et al. Oral microbial community composition is associated with pancreatic cancer: a case-control study in Iran. Cancer Med. 2020;9(2):797–806. doi:10.1002/cam4.2660.
  • Wei A-L, Mao L, Wei-Ming H, Zhen-Lu L, Yuan J, Liu H-Y, Zhou L-L, Li K, Ang L, Rosemary Fu M. et al. Oral microbiome and pancreatic cancer. WJG. 2020;9327(48):7679–7692. doi:10.3748/wjg.v26.i48.7679.
  • Nagata N, Nishijima S, Kojima Y, Hisada Y, Imbe K, Miyoshi-Akiyama T, Suda W, Kimura M, Aoki R, Sekine K. et al. Metagenomic identification of microbial signatures predicting pancreatic cancer from a multinational study. Gastroenterology. 2022;163(1):222–238. doi:10.1053/j.gastro.2022.03.054.
  • Kartal E, Schmidt TSB, Molina-Montes E, Rodríguez-Perales S, Wirbel J, Maistrenko OM, Akanni WA, Alashkar Alhamwe B, Alves RJ, Carrato A. et al. A faecal microbiota signature with high specificity for pancreatic cancer. Gut. 2022;71(7):1359–1372. doi:10.1136/gutjnl-2021-324755.
  • Lu H, Ren Z, Li A, Li J, Xu S, Zhang H, Jiang J, Yang J, Luo Q, Zhou K. et al. Tongue coating microbiome data distinguish patients with pancreatic head cancer from healthy controls. J Oral Microbiol. 2019;11(1):1563409. doi:10.1080/20002297.2018.1563409.
  • Fan X, Alekseyenko AV, Wu J, Peters BA, Jacobs EJ, Gapstur SM, Purdue MP, Abnet CC, Stolzenberg-Solomon R, Miller G. et al. Human oral microbiome and prospective risk for pancreatic cancer: a population-based nested case-control study. Gut. 2018;67(1):120–127. doi:10.1136/gutjnl-2016-312580.
  • Petrick JL, Wilkinson JE, Michaud DS, Cai Q, Gerlovin H, Signorello LB, Wolpin BM, Ruiz-Narváez EA, Long J, Yang Y. et al. The oral microbiome in relation to pancreatic cancer risk in African Americans. Br J Cancer. 2022;126(2):287–296. doi:10.1038/s41416-021-01578-5.
  • Michaud DS, Joshipura K, Giovannucci E, Fuchs CS. A prospective study of periodontal disease and pancreatic cancer in US male health professionals. J Natl Cancer Inst. 2007;99(2):171–175. doi:10.1093/jnci/djk021.
  • Yu J, Ploner A, Chen MS, Zhang J, Sandborgh-Englund G, Ye W. Poor dental health and risk of pancreatic cancer: a nationwide registry-based cohort study in Sweden, 2009–2016. Br J Cancer. 2022;127(12):2133–2140. doi:10.1038/s41416-022-02018-8.
  • Michaud DS, Izard J, Wilhelm-Benartzi CS, You D-H, Grote VA, Tjønneland A, Dahm CC, Overvad K, Jenab M, Fedirko V. et al. Plasma antibodies to oral bacteria and risk of pancreatic cancer in a large European prospective cohort study. Gut. 2013;62(12):1764–1770. doi:10.1136/gutjnl-2012-303006.
  • Huang J, Roosaar A, Axéll T, Ye W. A prospective cohort study on poor oral hygiene and pancreatic cancer risk. Int J Cancer. 2016;138(2):340–347. doi:10.1002/ijc.29710.
  • Chung L-M, Liang J-A, Lin C-L, Sun L-M, Kao C-H. Cancer risk in patients with candidiasis: a nationwide population-based cohort study. Oncotarget. 2017;8(38):63562–63573. doi:10.18632/oncotarget.18855.
  • Wei A, Zhao H, Cong X, Wang L, Chen Y, Gou J, Hu Z, Hu X, Tian Y, Li K. et al. Oral mycobiota and pancreatic ductal adenocarcinoma. BMC Cancer. 2022;22(1):1–13. doi:10.1186/s12885-022-10329-5.
  • Hall MW, Singh N, Ng KF, Lam DK, Goldberg MB, Tenenbaum HC, Neufeld JD, G Beiko R, Senadheera DB. Inter-personal diversity and temporal dynamics of dental, tongue, and salivary microbiota in the healthy oral cavity. Npj Biofilms Microbiomes. 2017;3(1):0–1. doi:10.1038/s41522-016-0011-0.
  • Wu J, Peters BA, Dominianni C, Zhang Y, Pei Z, Yang L, Ma Y, Purdue MP, Jacobs EJ, Gapstur SM. et al. Cigarette smoking and the oral microbiome in a large study of American adults. ISME J. 2016;10(10):2435–2446. doi:10.1038/ismej.2016.37.
  • Ren Z, Jiang J, Xie H, Li A, Lu H, Xu S, Zhou L, Zhang H, Cui G, Chen X. et al. Gut microbial profile analysis by MiSeq sequencing of pancreatic carcinoma patients in China. Oncotarget. 2017;8(56):95176–95191. doi:10.18632/oncotarget.18820.
  • Zhou W, Zhang D, Li Z, Jiang H, Li J, Ren R, Gao X, Li J, Wang X, Wang W. et al. The fecal microbiota of patients with pancreatic ductal adenocarcinoma and autoimmune pancreatitis characterized by metagenomic sequencing. J Transl Med. 2021;19(1):1–12. doi:10.1186/s12967-021-02882-7.
  • Half E, Keren N, Reshef L, Dorfman T, Lachter I, Kluger Y, Reshef N, Knobler H, Maor Y, Stein A. et al. Fecal microbiome signatures of pancreatic cancer patients. Sci Rep. 2019;9(1):1–12. doi:10.1038/s41598-019-53041-4.
  • Nejman D, Livyatan I, Fuks G, Gavert N, Zwang Y, Geller LT, Rotter-Maskowitz A, Weiser R, Mallel G, Gigi E. et al. The human tumor microbiome is composed of tumor type–specific intracellular bacteria. Science. 2020;368(6494):973–980. doi:10.1126/science.aay9189.
  • Del Castillo E, Meier R, Chung M, Koestler DC, Chen T, Paster BJ, Charpentier KP, Kelsey KT, Izard J, Michaud DS. et al. The microbiomes of pancreatic and duodenum tissue overlap and are highly subject specific but differ between pancreatic cancer and noncancer subjects. Cancer Epidemiol Biomarkers Prev. 2019;28(2):370–383. doi:10.1158/1055-9965.EPI-18-0542.
  • Aykut B, Pushalkar S, Chen R, Li Q, Abengozar R, Kim JI, Shadaloey SA, Wu D, Preiss P, Verma N. et al. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature. 2019;574(7777):264–267. doi:10.1038/s41586-019-1608-2.
  • Tan Q, Ma X, Yang B, Liu Y, Xie Y, Wang X, Yuan W, Ma J. Periodontitis pathogen porphyromonas gingivalis promotes pancreatic tumorigenesis via neutrophil elastase from tumor-associated neutrophils. Gut Microbes. 2022;14(1). doi:10.1080/19490976.2022.2073785.
  • Sethi V, Vitiello GA, Saxena D, Miller G, Dudeja V. The role of the microbiome in immunologic development and its implication for pancreatic cancer immunotherapy. Gastroenterology. 2019;156(7):2097–2115.e2. doi:10.1053/j.gastro.2018.12.045.
  • Diehl GE, Longman RS, Zhang J-X, Breart B, Galan C, Cuesta A, Schwab SR, Littman DR. Microbiota restricts trafficking of bacteria to mesenteric lymph nodes by CX 3 CR1 hi cells. Nature. 2013;494(7435):116–120. doi:10.1038/nature11809.
  • Thomas RM, Jobin C. Microbiota in pancreatic health and disease: the next frontier in microbiome research. Nat Rev Gastroenterol Hepatol. 2020;17(1):53–64. doi:10.1038/s41575-019-0242-7.
  • Ahuja M, Schwartz DM, Tandon M, Son A, Zeng M, Swaim W, Eckhaus M, Hoffman V, Cui Y, Xiao B. et al. Orai1-mediated antimicrobial secretion from pancreatic acini shapes the gut microbiome and regulates gut innate immunity. Cell Metab. 2017;25(3):635–646. doi:10.1016/j.cmet.2017.02.007.
  • Huang Y, Zhu N, Zheng X, Liu Y, Lu H, Yin X, Hao H, Tan Y, Wang D, Hu H. et al. Intratumor microbiome analysis identifies positive association between megasphaera and survival of Chinese patients with pancreatic ductal adenocarcinomas. Front Immunol. 2022;13:1–12. doi:10.3389/fimmu.2022.785422.
  • Mitsuhashi K, Nosho K, Sukawa Y, Matsunaga Y, Ito M, Kurihara H, Kanno S, Igarashi H, Naito T, Adachi Y. et al. Association of fusobacterium species in pancreatic cancer tissues with molecular features and prognosis. Oncotarget. 2015;6(9):7209–7220. doi:10.18632/oncotarget.3109.
  • Gaiser RA, Halimi A, Alkharaan H, Lu L, Davanian H, Healy K, Hugerth LW, Ateeb Z, Valente R, Fernández Moro C. et al. Enrichment of oral microbiota in early cystic precursors to invasive pancreatic cancer. Gut. 2019;68(12):2186–2194. doi:10.1136/gutjnl-2018-317458.
  • Riquelme E, Zhang Y, Zhang L, Montiel M, Zoltan M, Dong W, Quesada P, Sahin I, Chandra V, San Lucas A. et al. Tumor microbiome diversity and composition article tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell. 2019;178(4):795–806.e12. doi:10.1016/j.cell.2019.07.008.
  • Guo W, Zhang Y, Guo S, Mei Z, Liao H, Dong H, Wu K, Ye H, Zhang Y, Zhu Y. et al. Tumor microbiome contributes to an aggressive phenotype in the basal-like subtype of pancreatic cancer. Commun Biol. 2021;4(1):1019. doi:10.1038/s42003-021-02557-5.
  • Narunsky-Haziza L, Sepich-Poore GD, Livyatan I, Asraf O, Martino C, Nejman D, Gavert N, Stajich JE, Amit G, González A. et al. Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell. 2022;185(20):3789–3806.e17. doi:10.1016/j.cell.2022.09.005.
  • Alam A, Levanduski E, Denz P, Villavicencio HS, Bhatta M, Alhorebi L, Zhang Y, Gomez EC, Morreale B, Senchanthisai S. et al. Article fungal mycobiome drives IL-33 secretion and type 2 immunity in pancreatic cancer ll article fungal mycobiome drives IL-33 secretion and type 2 immunity in pancreatic cancer. Cancer Cell. 2022;40(2):153–167.e11. doi:10.1016/j.ccell.2022.01.003.
  • Eckhoff AM, Fletcher AA, Kelly MS, Dohlman A, McIntyre CA, Shen X, Iyer MK, Nussbaum DP, Allen PJ. Comprehensive assessment of the intrinsic pancreatic microbiome. bioRxiv. 2023;2023(8):12.553074. doi:10.1101/2023.08.12.553074.
  • Poore GD, Kopylova E, Zhu Q, Carpenter C, Fraraccio S, Wandro S, Kosciolek T, Janssen S, Metcalf J, Song SJ. et al. Microbiome analyses of blood and tissues suggest cancer diagnostic approach. Nature. 2020;579(7800):567–574. doi:10.1038/s41586-020-2095-1.
  • Gihawi A, Ge Y, Lu J, Puiu D, Xu A, Cooper CS, Brewer DS, Pertea M, Salzberg SL. Major data analysis errors invalidate cancer microbiome findings. Am Soc Microbiol. 2023;14(5):e01607–23. doi:10.1128/mbio.01607-23.
  • Fletcher AA, Kelly MS, Eckhoff AM, Allen PJ. Revisiting the intrinsic mycobiome in pancreatic cancer. Nature. 2023;620(7972):E1–E6. doi:10.1038/s41586-023-06292-1.
  • Rubinstein MR, Wang X, Liu W, Hao Y, Cai G, Han Y. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-Cadherin/β-catenin signaling via its FadA Adhesin. Cell Host Microbe. 2013;14(2):195–206. doi:10.1016/j.chom.2013.07.012.
  • Bullman S, Pedamallu CS, Sicinska E, Clancy TE, Zhang X, Cai D, Neuberg D, Huang K, Guevara F, Nelson T. et al. Analysis of fusobacterium persistence and antibiotic response in colorectal cancer. Science. 2017;358(6369):1443–1448. doi:10.1126/science.aal5240.
  • Amieva M, Peek RM. Pathobiology of Helicobacter pylori–induced gastric cancer. Gastroenterology. 2016;150(1):64–78. doi:10.1053/j.gastro.2015.09.004.
  • Udayasuryan B, Ahmad RN, Nguyen TT&, Umaña A, Monét Roberts L, Sobol P, Jones SD, Munson JM, Slade DJ, Verbridge SS. et al. Fusobacterium nucleatum induces proliferation and migration in pancreatic cancer cells through host autocrine and paracrine signaling. Sci Signal. 2022;15(756):eabn4948. doi:10.1126/scisignal.abn4948.
  • Coker OO, Nakatsu G, Dai RZ, Wu WKK, Wong SH, Ng SC, Chan FKL, Sung JJY, Yu J. Enteric fungal microbiota dysbiosis and ecological alterations in colorectal cancer. Gut. 2019;68(4):654–662. doi:10.1136/gutjnl-2018-317178.
  • Sam QH, Chang MW, Chai LYA. The fungal mycobiome and its interaction with gut bacteria in the host. Int J Mol Sci. 2017;18(2):330. doi:10.3390/ijms18020330.
  • Iida N, Dzutsev A, Stewart CA, Smith L, Bouladoux N, Weingarten RA, Molina DA, Salcedo R, Back T, Cramer S. et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science. 2013;342(6161):967–970. doi:10.1126/science.1240527.
  • Alexander JL, Wilson ID, Teare J, Marchesi JR, Nicholson JK, Kinross JM. Gut microbiota modulation of chemotherapy efficacy and toxicity. Nat Rev Gastroenterol Hepatol. 2017;14(6):356–365. doi:10.1038/nrgastro.2017.20.
  • García-González AP, Ritter AD, Shrestha S, Andersen EC, Yilmaz LS, Walhout AJM. Bacterial metabolism affects the C. elegans response to cancer chemotherapeutics. Cell. 2017;169(3):431–441.e8. doi:10.1016/j.cell.2017.03.046.
  • Scott TA, Quintaneiro LM, Norvaisas P, Lui PP, Wilson MP, Leung K-Y, Herrera-Dominguez L, Sudiwala S, Pessia A, Clayton PT. et al. Host-miCrobe co-metabolism dictates cancer drug efficacy in C. elegans. Cell. 2017;169(3):442–456.e18. doi:10.1016/j.cell.2017.03.040.
  • Yu TC, Guo F, Yu Y, Sun T, Ma D, Han J, Qian Y, Kryczek I, Sun D, Nagarsheth N. et al. Fusobacterium nucleatum promotes chemoresistance to colorectal cancer by modulating autophagy. Cell. 2017;170(3):548–563.e16. doi:10.1016/j.cell.2017.07.008.
  • Zhang S, Yang Y, Weng W, Guo B, Cai G, Ma Y, Cai S. Fusobacterium nucleatum promotes chemoresistance to 5-fluorouracil by upregulation of BIRC3 expression in colorectal cancer. J Exp Clin Cancer Res. 2019;38(1):1–13. doi:10.1186/s13046-018-0985-y.
  • Imai H, Saijo K, Komine K, Yoshida Y, Sasaki K, Suzuki A, Ouchi K, Takahashi M, Takahashi S, Shirota H. et al. Antibiotics improve the treatment efficacy of oxaliplatin-based but not irinotecan-based therapy in advanced colorectal cancer patients. J Oncol. 2020;2020:1–8. doi:10.1155/2020/1701326.
  • Yuan L, Zhang S, Li H, Yang F, Mushtaq N, Ullah S, Shi Y, An C, Xu J. The influence of gut microbiota dysbiosis to the efficacy of 5-fluorouracil treatment on colorectal cancer. Biomed Pharmacother. 2018;108:184–193. doi:10.1016/j.biopha.2018.08.165.
  • Roberti MP, Yonekura S, Duong CPM, Picard M, Ferrere G, Tidjani Alou M, Rauber C, Iebba V, Lehmann CHK, Amon L. et al. Chemotherapy-induced ileal crypt apoptosis and the ileal microbiome shape immunosurveillance and prognosis of proximal colon cancer. Nat Med. 2020;26(6):919–931. doi:10.1038/s41591-020-0882-8.
  • Fulop DJ, Zylberberg HM, Wu YL, Aronson A, Labiner AJ, Wisnivesky J, Cohen DJ, Sigel KM, Lucas AL. Association of antibiotic receipt with survival among patients with metastatic pancreatic ductal adenocarcinoma receiving chemotherapy. JAMA Netw Open. 2023;6(3):E234254. doi:10.1001/jamanetworkopen.2023.4254.
  • Imai H, Saijo K, Komine K, Otsuki Y, Ohuchi K, Sato Y, Okita A, Takahashi M, Takahashi S, Shirota H. et al. Antibiotic therapy augments the efficacy of gemcitabine-containing regimens for advanced cancer: a retrospective study. Cancer Manag Res. 2019;11:7953–7965. doi:10.2147/CMAR.S215.697.
  • Yue B, Gao R, Wang Z, Dou W. Microbiota-host-irinotecan axis: a new insight toward irinotecan chemotherapy. Front Cell Infect Microbiol. 2021;11:1–15. doi:10.3389/fcimb.2021.710945.
  • Wallace BD, Wang H, Lane KT, Scott JE, Orans J, Koo JS, Venkatesh M, Jobin C, Yeh L-A, Mani S. et al. Alleviating cancer drug toxicity by inhibiting a bacterial enzyme. Science. 2010;330(6005):831–835. doi:10.1126/science.1191175.
  • Lin HY, Chen C-Y, Lin T-C, Yeh L-F, Hsieh W-C, Gao S, Burnouf P-A, Chen B-M, Hsieh T-J, Dashnyam P. et al. Entropy-driven binding of gut bacterial β-glucuronidase inhibitors ameliorates irinotecan-induced toxicity. Commun Biol. 2021;4(1):1–10. doi:10.1038/s42003-021-01815-w.
  • Pedroso SHSP, Vieira AT, Bastos RW, Oliveira JS, Cartelle CT, Arantes RME, Soares PMG, Generoso SV, Cardoso VN, Teixeira MM. et al. Evaluation of mucositis induced by irinotecan after microbial colonization in germ-free mice. Microbiology. 2015;161(10):1950–1960. doi:10.1099/mic.0.000149.
  • Kehrer DFS, Sparreboom A, Verweij J, de Bruijn P, Nierop CA, van de Schraaf J, Ruijgrok EJ, de Jonge MJ. Advances in brief modulation of irinotecan-induced diarrhea by cotreatment with neomycin in cancer patients. Clin Cancer Res. 2001;7:1136–1141.
  • de Jong FA, Kehrer DFS, Mathijssen RHJ, Creemers G-J, de Bruijn P, van Schaik RHN, Planting AST, van der Gaast A, Eskens FALM, Janssen JTP. et al. Prophylaxis of irinotecan-induced diarrhea with neomycin and potential role for UGT1A1*28 genotype screening: a double-blind, randomized, placebo-controlled study. Oncologist. 2006;11(8):944–954. doi:10.1634/theoncologist.11-8-944.
  • Sharma R, Tobin P, Clarke SJ. Management of chemotherapy-induced nausea, vomiting, oral mucositis, and diarrhoea. Lancet Oncol. 2005;6(2):93–102. doi:10.1016/S1470-2045(05)01735-3.
  • Makihara K, Nakamura S, Miyagi K, Ueno H, Nakata I. Clarithromycin co-administration does not increase irinotecan (CPT-11) toxicity in colorectal cancer patients. Cancer Chemother Pharmacol. 2017;80(3):527–533. doi:10.1007/s00280-017-3388-4.
  • Kurita A, Kado S, Matsumoto T, Asakawa N, Kaneda N, Kato I, Uchida K, Onoue M, Yokokura T. Streptomycin alleviates irinotecan-induced delayed-onset diarrhea in rats by a mechanism other than inhibition of β-glucuronidase activity in intestinal lumen. Cancer Chemother Pharmacol. 2011;67(1):201–213. doi:10.1007/s00280-010-1310-4.
  • Lo EKK, Leung HKM, Zhang F, El-Nezami H. Gut microbiota: Impact on 5-fluorouracil efficacy and toxicity. Curr Opin Toxicol. 2023;36:100423. doi:10.1016/j.cotox.2023.100423.
  • Shen S, Lim G, You Z, Ding W, Huang P, Ran C, Doheny J, Caravan P, Tate S, Hu K. et al. Gut microbiota is critical for the induction of chemotherapy-induced pain. Nat Neurosci. 2017;20(9):1213–1216. doi:10.1038/nn.4606.
  • Panebianco C, Adamberg K, Jaagura M, Copetti M, Fontana A, Adamberg S, Kolk K, Vilu R, Andriulli A, Pazienza V. et al. Influence of gemcitabine chemotherapy on the microbiota of pancreatic cancer xenografted mice. Cancer Chemother Pharmacol. 2018;81(4):773–782. doi:10.1007/s00280-018-3549-0.
  • Hamouda N, Sano T, Oikawa Y, Ozaki T, Shimakawa M, Matsumoto K, Amagase K, Higuchi K, Kato S. Apoptosis, dysbiosis and expression of inflammatory cytokines are sequential events in the development of 5-fluorouracil-induced intestinal mucositis in mice. Basic Clin Pharmacol Toxicol. 2017;121(3):159–168. doi:10.1111/bcpt.12793.
  • Goel N, Nadler A, Reddy S, Hoffman JP, Pitt HA. Biliary microbiome in pancreatic cancer: alterations with neoadjuvant therapy. Hpb. 2019;21(12):1753–1760. doi:10.1016/j.hpb.2019.04.005.
  • Nadeem SO, Jajja MR, Maxwell DW, Pouch SM, Sarmiento JM. Neoadjuvant chemotherapy for pancreatic cancer and changes in the biliary microbiome. Am J Surg. 2021;222(1):3–7. doi:10.1016/j.amjsurg.2020.09.042.
  • Balachandran VP, Łuksza M, Zhao JN, Makarov V, Moral JA, Remark R, Herbst B, Askan G, Bhanot U, Senbabaoglu Y. et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature. 2017;551(7681):S512–S516. doi:10.1038/nature24462.
  • Vétizou M, Pitt JM, Daillère R, Lepage P, Waldschmitt N, Flament C, Rusakiewicz S, Routy B, Roberti MP, Duong CPM. et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science. 2015;350(6264):1079–1084. doi:10.1126/science.aad1329.
  • Sivan A, Corrales L, Hubert N, Williams JB, Aquino-Michaels K, Earley ZM, Benyamin FW, Man Lei Y, Jabri B, Alegre M-L. et al. Commensal bifidobacterium promotes antitumor immunity and facilitates anti–PD-L1 efficacy. Science. 2015;350(6264):1084–1089. doi:10.1126/science.aac4255.
  • Routy B, Le Chatelier E, Derosa L, Duong CPM, Alou MT, Daillère R, Fluckiger A, Messaoudene M, Rauber C, Roberti MP. et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97. doi:10.1126/science.aan3706.
  • Si W, Liang H, Bugno J, Xu Q, Ding X, Yang K, Fu Y, Weichselbaum RR, Zhao X, Wang L. et al. Lactobacillus rhamnosus GG induces cGAS/STING- dependent type i interferon and improves response to immune checkpoint blockade. Gut. 2022;71(3):521–533. doi:10.1136/gutjnl-2020-323426.
  • Tanoue T, Morita S, Plichta DR, Skelly AN, Suda W, Sugiura Y, Narushima S, Vlamakis H, Motoo I, Sugita K. et al. A defined commensal consortium elicits CD8 T cells and anti-cancer immunity. Nature. 2019;565(7741):600–605. doi:10.1038/s41586-019-0878-z.
  • Davar D, Dzutsev AK, McCulloch JA, Rodrigues RR, Chauvin J-M, Morrison RM, Deblasio RN, Menna C, Ding Q, Pagliano O. et al. Fecal microbiota transplant overcomes resistance to anti–PD-1 therapy in melanoma patients. Science. 2021;371(6529):595–602. doi:10.1126/science.abf3363.
  • Baruch EN, Youngster I, Ben-Betzalel G, Ortenberg R, Lahat A, Katz L, Adler K, Dick-Necula D, Raskin S, Bloch N. et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science. 2021;371(6529):602–609. doi:10.1126/science.abb5920.
  • Postler TS, Ghosh S. Understanding the holobiont: how microbial metabolites affect human health and shape the immune system. Cell Metab. 2017;26(1):110–130. doi:10.1016/j.cmet.2017.05.008.
  • Natoni F, Diolordi L, Santoni C, Gilardini Montani MS. Sodium butyrate sensitises human pancreatic cancer cells to both the intrinsic and the extrinsic apoptotic pathways. Biochim Biophys Acta - Mol Cell Res. 2005;1745(3):318–329. doi:10.1016/j.bbamcr.2005.07.003.
  • Bülow R, Fitzner B, Sparmann G, Emmrich J, Liebe S, Jaster R. Antifibrogenic effects of histone deacetylase inhibitors on pancreatic stellate cells. Biochem Pharmacol. 2007;74(12):1747–1757. doi:10.1016/j.bcp.2007.08.023.
  • Panebianco C, Villani A, Pisati F, Orsenigo F, Ulaszewska M, Latiano TP, Potenza A, Andolfo A, Terracciano F, Tripodo C. et al. Butyrate, a postbiotic of intestinal bacteria, affects pancreatic cancer and gemcitabine response in in vitro and in vivo models. Biomed Pharmacother. 2022;151:113163. doi:10.1016/j.biopha.2022.113163.
  • Shuwen H, Yangyanqiu W, Jian C, Boyang H, Gong C, Jing Z. Synergistic effect of sodium butyrate and oxaliplatin on colorectal cancer. Transl Oncol. 2023;27:101598. doi:10.1016/j.tranon.2022.101598.
  • Yue X, Wen S, Long-Kun D, Man Y, Chang S, Min Z, Shuang-Yu L, Xin Q, Jie M, Liang W. et al. Three important short-chain fatty acids (SCFAs) attenuate the inflammatory response induced by 5-FU and maintain the integrity of intestinal mucosal tight junction. BMC Immunol. 2022;23(1):1–13. doi:10.1186/s12865-022-00495-3.
  • Gao J, Xu K, Liu H, Liu G, Bai M, Peng C, Li T, Yin Y. Impact of the gut microbiota on intestinal immunity mediated by tryptophan metabolism. Front Cell Infect Microbiol. 2018;8:1–22. doi:10.3389/fcimb.2018.00013.
  • Hezaveh K, Shinde RS, Klötgen A, Halaby MJ, Lamorte S, Ciudad MT, Quevedo R, Neufeld L, Liu ZQ, Jin R. et al. Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity. 2022;55(2):324–340.e8. doi:10.1016/j.immuni.2022.01.006.
  • Collins SL, Stine JG, Bisanz JE, Okafor CD, Patterson AD. Bile acids and the gut microbiota: metabolic interactions and impacts on disease. Nat Rev Microbiol. 2023;21(4):236–247. doi:10.1038/s41579-022-00805-x.
  • Yang C, Yuan H, Gu J, Xu D, Wang M, Qiao J, Yang X, Zhang J, Yao M, Gu J. et al. ABCA8-mediated efflux of taurocholic acid contributes to gemcitabine insensitivity in human pancreatic cancer via the S1PR2-ERK pathway. Cell Death Discov. 2021;7(1). doi:10.1038/s41420-020-00390-z.
  • Gál E, Veréb Z, Kemény L, Rakk D, Szekeres A, Becskeházi E, Tiszlavicz L, Takács T, Czakó L, Hegyi P. et al. Bile accelerates carcinogenic processes in pancreatic ductal adenocarcinoma cells through the overexpression of MUC4. Sci Rep. 2020;10(1):1–16. doi:10.1038/s41598-020-79181-6.
  • Tucker ON, Dannenberg AJ, Yang EK, Fahey TJ. Bile acids induce cyclooxygenase-2 expression in human pancreatic cancer cell lines. Carcinogenesis. 2004;25(3):419–423. doi:10.1093/carcin/bgh010.
  • Nagathihalli NS, Beesetty Y, Lee W, Washington MK, Chen X, Lockhart AC, Merchant NB. Novel mechanistic insights into ectodomain shedding of egfr ligands amphiregulin and TGF-α: impact on gastrointestinal cancers driven by secondary bile acids. Cancer Res. 2014;74(7):2062–2072. doi:10.1158/0008-5472.CAN-13-2329.
  • Shrader HR, Miller AM, Tomanek-Chalkley A, McCarthy A, Coleman KL, Ear PH, Mangalam AK, Salem AK, Chan CHF. Effect of bacterial contamination in bile on pancreatic cancer cell survival. Surgery. 2021;169(3):617–622. doi:10.1016/j.surg.2020.09.029.
  • Mendez R, Kesh K, Arora N, Di Martino L, McAllister F, Merchant N, Banerjee S, Banerjee S. Microbial dysbiosis and polyamine metabolism as predictive markers for early detection of pancreatic cancer. Carcinogenesis. 2020;41(5):561–570. doi:10.1093/carcin/bgz116.
  • Nakkina SP, Gitto SB, Pandey V, Parikh JG, Geerts D, Maurer HC, Olive KP, Phanstiel O, Altomare DA. Differential expression of polyamine pathways in human pancreatic tumor progression and effects of polyamine blockade on tumor microenvironment. Cancers. 2021;13(24):6391. doi:10.3390/cancers13246391.
  • Lee M, Dennis C, Naqvi I, Dailey L, Lorzadeh A, Ye G, Zaytouni T, Adler A, Hitchcock DS, Lin L. et al. Ornithine aminotransferase supports polyamine synthesis in pancreatic cancer. Nature. 2023;616(7956):339–347. doi:10.1038/s41586-023-05891-2.
  • Tofalo R, Cocchi S, Suzzi G. Polyamines and gut microbiota. Front Nutr. 2019;6:1–5. doi:10.3389/fnut.2019.00016.
  • Yin J, Ren W, Huang X, Deng J, Li T, Yin Y. Potential mechanisms connecting purine metabolism and cancer therapy. Front Immunol. 2018;9:1–8. doi:10.3389/fimmu.2018.01697.
  • Santana-Codina N, Roeth AA, Zhang Y, Yang A, Mashadova O, Asara JM, Wang X, Bronson RT, Lyssiotis CA, Ying H. et al. Oncogenic KRAS supports pancreatic cancer through regulation of nucleotide synthesis. Nat Commun. 2018;9(1):4945. doi:10.1038/s41467-018-07472-8.
  • Lee JS, Wang RX, Goldberg MS, Clifford GP, Kao DJ, Colgan SP. Microbiota-sourced purines support wound healing and mucous barrier function. iScience. 2020;23(6):101226. doi:10.1016/j.isci.2020.101226.
  • Mager LF, Burkhard R, Pett N, Cooke NCA, Brown K, Ramay H, Paik S, Stagg J, Groves RA, Gallo M. et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy. Science. 2020;369(6510):1481–1489. doi:10.1126/science.abc3421.
  • Capurso G, Sette C. Drug resistance in pancreatic cancer: new player caught in act. EBioMedicine. 2019;40:39–40. doi:10.1016/j.ebiom.2019.02.008.
  • Plagemann CN, Hidalgo M, Garrido-Laguna I. From state- of-the- art treatments to novel therapies for advanced-stage pancreatic cancer. Nat Rev Clin Oncol. 2020;17(2):108–123. doi:10.1038/s41571-019-0281-6.
  • Zmora N, Suez J, Elinav E. You are what you eat: diet, health and the gut microbiota. Nat Rev Gastroenterol Hepatol. 2019;16(1):35–56. doi:10.1038/s41575-018-0061-2.
  • Yang J, Wei H, Zhou Y, Szeto C-H, Li C, Lin Y, Coker OO, Lau HCH, Chan AWH, Sung JJY. et al. High-fat diet promotes colorectal tumorigenesis through modulating gut microbiota and metabolites. Gastroenterology. 2022;162(1):135–149.e2. doi:10.1053/j.gastro.2021.08.041.
  • Bultman SJ. Molecular pathways: gene–environment interactions regulating dietary fiber induction of proliferation and apoptosis via butyrate for cancer prevention. Clin Cancer Res. 2014;20(4):799–803. doi:10.1158/1078-0432.CCR-13-2483.
  • Mao QQ, Lin Y-W, Chen H, Qin J, Zheng X-Y, Xu X, Xie L-P. Dietary fiber intake is inversely associated with risk of pancreatic cancer: a meta-analysis. Asia Pac J Clin Nutr. 2017;26(1):89–96. doi:10.6133/apjcn.102015.03.
  • Panebianco C, Adamberg K, Adamberg S, Saracino C, Jaagura M, Kolk K, Di Chio A, Graziano P, Vilu R, Pazienza V. et al. Engineered resistant-starch (ERS) diet shapes colon microbiota profile in parallel with the retardation of tumor growth in in vitro and in vivo pancreatic cancer models. Nutrients. 2017;9(4):331. doi:10.3390/nu9040331.
  • D’Aronzo M, Vinciguerra M, Mazza T, Panebianco C, Saracino C, Pereira SP, Graziano P, Pazienza V. Fasting cycles potentiate the efficacy of gemcitabine treatment in in vitro and in vivo pancreatic cancer models. Oncotarget. 2015;6(21):18545–18557. doi:10.18632/oncotarget.4186.
  • Lin XB, Farhangfar A, Valcheva R, Sawyer MB, Dieleman L, Schieber A, Gänzle MG, Baracos V. The role of intestinal microbiota in development of irinotecan toxicity and in toxicity reduction through dietary fibres in rats. PloS One. 2014;9(1):e83644. doi:10.1371/journal.pone.0083644.
  • Mirmiran P, Bahadoran Z, Gaeini Z. Common limitations and challenges of dietary clinical trials for translation into clinical practices. Int J Endocrinol Metab. 2021;19(3):1–9. doi:10.5812/ijem.108170.
  • Fu X, Liu Z, Zhu C, Mou H, Kong Q. Nondigestible carbohydrates, butyrate, and butyrate-producing bacteria. Crit Rev Food Sci Nutr. 2019;59(sup1):S130–S152. doi:10.1080/10408398.2018.1542587.
  • Panebianco C, Pisati F, Ulaszewska M, Andolfo A, Villani A, Federici F, Laura M, Rizzi E, Potenza A, Latiano TP. et al. Tuning gut microbiota through a probiotic blend in gemcitabine‐treated pancreatic cancer xenografted mice. Clin Transl Med. 2021;11(11):1–7. doi:10.1002/ctm2.580.
  • Prajapati N, Patel J, Singh S, Yadav VK, Joshi C, Patani A, Prajapati D, Sahoo DK, Patel A. Postbiotic production: harnessing the power of microbial metabolites for health applications. Front Microbiol. 2023;14:1–16. doi:10.3389/fmicb.2023.1306192.
  • Chrysostomou D, Roberts LA, Marchesi JR, Kinross JM. Gut microbiota modulation of efficacy and toxicity of cancer chemotherapy and immunotherapy. Gastroenterology. 2023;164(2):198–213. doi:10.1053/j.gastro.2022.10.018.
  • Mohindroo C, Hasanov M, Rogers JE, Dong W, Prakash LR, Baydogan S, Mizrahi JD, Overman MJ, Varadhachary GR, Wolff RA. et al. Antibiotic use influences outcomes in advanced pancreatic adenocarcinoma patients. Cancer Med. 2021;10(15):5041–5050. doi:10.1002/cam4.3870.
  • Hwang YJ, Myung H. Engineered bacteriophage T7 as a potent anticancer agent in vivo. Front Microbiol. 2020;11:491001. doi:10.3389/fmicb.2020.491001.
  • Kabwe M, Dashper S, Tucci J. The microbiome in pancreatic cancer-implications for diagnosis and precision bacteriophage therapy for this low survival disease. Front Cell Infect Microbiol. 2022;12:1–14. doi:10.3389/fcimb.2022.871293.
  • Identifier: NCT04975217. Fecal microbial transplants for the treatment of pancreatic cancer. US National Library Of Medicine. (Accessed: 20th October 2023). https://classic.clinicaltrials.gov/ct2/show/NCT04975217