Next Article in Journal
Deep Learning Model-Based Architectures for Lung Tumor Mutation Profiling: A Systematic Review
Previous Article in Journal
Impact of Dosimetric Parameters on Tumor Control in Stereotactic Radiotherapy for Pancreatic Cancer: A Prospective Study on 104 Patients Treated with Simultaneous Integrated Protection (SIP)
Previous Article in Special Issue
Cell Line-Dependent Internalization, Persistence, and Immunomodulatory Effects of Staphylococcus aureus in Triple-Negative Breast Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microbiota and Pancreatic Cancer: New Therapeutic Frontiers Between Engineered Microbes, Metabolites and Non-Bacterial Components

by
Sara Sofia De Lucia
1,
Enrico Celestino Nista
1,2,
Marcello Candelli
1,2,
Sebastiano Archilei
1,
Franziska Deutschbein
1,
Enrico Capuano
1,
Antonio Gasbarrini
1,
Francesco Franceschi
1,2 and
Giulia Pignataro
1,2,*
1
Department of Medical and Surgery Sciences, Università Cattolica del Sacro Cuore di Roma, 00168 Roma, Italy
2
Department of Emergency, Anesthesiologic and Reanimation Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(22), 3618; https://doi.org/10.3390/cancers17223618
Submission received: 16 October 2025 / Revised: 4 November 2025 / Accepted: 9 November 2025 / Published: 10 November 2025
(This article belongs to the Special Issue New Insights into Microbial-Based Cancer Therapy)

Simple Summary

Pancreatic ductal adenocarcinoma is one of the deadliest forms of cancer, with survival rates that have changed little over time. Traditional treatments are often ineffective, highlighting the urgent need for new strategies. Recent studies have shown that microorganisms living in the body (such as bacteria, fungi, and viruses) can influence the development and response of pancreatic cancer to therapy. This review examines the impact of these microbial communities on tumor growth, the immune system, and the efficacy of current treatments. It also examines innovative therapeutic approaches that use beneficial microbes or target harmful ones, including probiotics, engineered bacteria, bacteriophages, and fecal microbiota transplantation. With a better understanding and manipulation of the relationship between the microbiome and cancer, researchers may discover new ways to diagnose, prevent, and treat pancreatic cancer more effectively.

Abstract

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most aggressive and lethal human malignancies, with five-year survival rates showing only marginal improvement despite decades of intensive research. Its dismal prognosis reflects a combination of intrinsic biological aggressiveness, late clinical presentation, and marked resistance to standard therapies, underscoring the urgent need for innovative diagnostic and therapeutic approaches. Growing evidence indicates that the microbiome is a modifiable factor influencing the onset, progression, and treatment response of PDAC. Microbial communities originating from the gut, oral cavity, and even the tumor microenvironment can shape carcinogenic pathways, modulate immune activity, and alter the efficacy of chemotherapy and immunotherapy. In addition to bacteria, fungal and viral populations are emerging as relevant contributors within this complex ecosystem. This review provides a comprehensive overview of the current mechanistic and translational evidence linking the microbiome to PDAC biology and therapy. It further explores microbiota-targeted interventions—such as probiotics, postbiotics, engineered bacterial strains, bacteriophages, oncolytic viruses, and fecal microbiota transplantation—as promising adjuncts to conventional treatments. A deeper understanding of host–microbiome interactions could yield novel biomarkers and open innovative avenues for precision medicine in PDAC, ultimately improving patient outcomes and reshaping therapeutic paradigms. Integrating microbiome-based strategies into PDAC management may thus represent a crucial step toward more effective and personalized oncologic care.

1. Introduction

The human gut microbiota constitutes a highly diverse ecological community of bacteria, fungi, viruses, and parasites, collectively comprising approximately 100 trillion microorganisms that play a pivotal role in maintaining host health [1]. Once considered a passive colonizer, the microbiota is a dynamic regulator of host physiology, orchestrating immune maturation, nutrient and drug metabolism, and the maintenance of mucosal integrity [2,3]. Disruptions in microbial composition or diversity have been associated with a wide range of pathological conditions, including malignancies [4,5]. The concept of bidirectional communication between the gut and distant organs is well established, with the gut–brain axis serving as the classical model [6]. More recently, this paradigm has been expanded to include a gut–pancreas axis, highlighting the reciprocal interactions between these systems. Pancreatic secretions—comprising digestive enzymes and antimicrobial peptides—can influence intestinal microbial communities, whereas microbial dysbiosis and subsequent translocation across the gut barrier may trigger pancreatic inflammation [7]. This bidirectional relationship is particularly relevant in the context of pancreatic ductal adenocarcinoma (PDAC), the most prevalent pancreatic malignancy and the sixth leading cause of cancer-related mortality worldwide [8,9]. Despite significant advances in oncology, the prognosis for PDAC remains poor, with five-year survival rates showing minimal improvement due to late-stage presentation and the absence of effective screening strategies [8]. Established risk factors include cigarette smoking, chronic pancreatitis, diabetes mellitus, family history, alcohol consumption, obesity, and inherited predisposition [10]. However, these factors alone cannot fully explain the onset of disease, suggesting the involvement of additional contributors. Among these, the microbiome has emerged as a potential key determinant [10]. Increasing evidence implicates microbial communities, including those from the gut, oral cavity, and intratumoral niches, in the initiation and progression of PDAC [10]. Dysbiosis has been linked not only to carcinogenesis but also to the remodeling of the tumor microenvironment, thereby modulating immune regulation and therapeutic efficacy [11]. Intratumoral microorganisms, in particular, may affect responsiveness to chemotherapy and immunotherapy, opening the door to precision-based therapeutic approaches [12]. This review synthesizes current evidence on the intricate interplay between the microbiota and pancreatic cancer, with a focus on dysbiosis, microbial metabolites, and the emerging role of non-bacterial components. It further explores how host–microbe interactions influence the tumor microenvironment and therapeutic response and discusses innovative microbiome-centered strategies—including microbiota editing and engineered microbial consortia—as potential supplements to existing therapies.

2. Materials and Methods

Relevant publications were identified through comprehensive searches of the PubMed, Scopus, and Embase databases. The search strategy combined Medical Subject Headings (MeSH) terms and free-text keywords, including “pancreatic cancer,” “pancreatic ductal adenocarcinoma,” “microbiota,” “gut microbiota,” “dysbiosis,” “oral microbiota,” “periodontal disease,” “fecal microbiota transplantation,” “probiotics,” “prebiotics,” “antibiotics,” and “microbial metabolites.” Boolean operators (“AND,” “OR”) were used to refine or broaden the search appropriately. The search was limited to peer-reviewed English-language articles published within the past 20 years. Following the removal of duplicates, all retrieved records were screened for relevance. Titles and abstracts were assessed for eligibility, with full-text review conducted when preliminary screening suggested potential inclusion. Studies were eligible if they examined any aspect of the relationship between the microbiota (intestinal, oral, or other body sites) and pancreatic ductal adenocarcinoma (PDAC), including mechanistic insights, diagnostic or prognostic implications, and therapeutic relevance. Both original research and review articles were included to ensure a broad and balanced synthesis of available evidence. The methodological quality and scientific rigor of each study were evaluated, with particular attention to study design, sample size, analytical techniques, and relevance to the research objectives. Data from eligible studies was narratively synthesized to identify convergent findings, clarify discrepancies and current knowledge gaps, and outline emerging trends in microbiota–PDAC research.

3. Oncobiosis and Microbial Metabolite Signaling in Pancreatic Adenocarcinoma

3.1. Oral and Oral–Gut Microbiota Axis in the Pathogenesis and Progression of Pancreatic Adenocarcinoma

The oral cavity hosts a complex microbial ecosystem comprising thousands of microorganisms collectively known as the oral microbiota [13]. Increasing evidence indicates that these microbial species are essential for maintaining oral homeostasis but may also contribute to oncogenic processes, particularly under conditions of dysbiosis [14]. Oral health is closely tied to the composition of this microbiome; for example, periodontal disease—an inflammatory condition marked by immune dysregulation—can result from alterations in the oral microbial community [15]. Oral dysbiosis has been associated with a variety of systemic diseases, including diabetes, atherosclerosis, and cancer [16]. Multiple lines of evidence suggest that periodontal pathogens are linked to the development of several malignancies, including pancreatic cancer [17]. Notably, periodontal disease is now recognized as an independent risk factor for pancreatic cancer [18]. In a prospective study, elevated serum antibodies against Porphyromonas gingivalis (P. gingivalis) ATCC 53978 were detected years before diagnosis. They were significantly more prevalent among individuals who later developed pancreatic cancer, suggesting that oral dysbiosis may predispose to this malignancy even up to five years prior to its onset [19]. Moreover, P. gingivalis has been identified in pancreatic lesions in murine models and shown to induce pancreatic acinar-to-ductal metaplasia in vitro [20]. In addition, the dynamic interplay between oral and gut microbial communities—referred to as the “oral–gut microbiota axis”—has been shown to contribute to the pathogenesis of pancreatic ductal adenocarcinoma (PDAC) through digestive and circulatory translocation toward the pancreatic gland [21]. Finally, a recent review by Papa et al. reported that several oral species, including Neisseria elongata, Streptococcus mitis, Granulicatella adiacens, and Aggregatibacter actinomycetemcomitans, are associated with an increased risk of pancreatic cancer [22]. Although the underlying pathogenic mechanisms remain incompletely elucidated, these findings open promising avenues for early diagnosis and risk stratification, as well as for the development of innovative microbiome-based therapeutic strategies.

3.2. Role of the Gut Microbiota in the Pathogenesis and Progression of Pancreatic Adenocarcinoma

The gut microbiota is increasingly recognized as a key factor in pancreatic tumorigenesis and disease progression. In a prospective study, Nagata et al. analyzed fecal samples from patients with pancreatic cancer and compared them with those from healthy controls [23]. They reported an enrichment of Veillonella spp. and Actinomyces spp., along with a reduction in Clostridiales—including unclassified Lachnospiraceae, Eubacterium ventriosum, and Faecalibacterium prausnitzii—particularly among individuals with advanced disease [23]. A subsequent metagenomic shotgun sequencing study examined the association between single-nucleotide polymorphisms (SNPs) within gut bacterial genomes and pancreatic cancer. Among the analyzed taxa, Lachnospiraceae exhibited the highest SNP density, particularly in patients with tumors located in the pancreatic head, where SNP abundance was inversely correlated with pancreatic cancer risk [24]. The role of Helicobacter pylori (H. pylori) in pancreatic carcinogenesis remains a topic of controversy. While a recent study found no association between H. pylori seropositivity and pancreatic cancer risk [25], earlier reports suggested a positive link [26]. Thus, the contribution of H. pylori to pancreatic tumorigenesis remains unresolved. Beyond bacteria, fungal communities may also participate in pancreatic oncogenesis. In particular, Malassezia species have been identified as more abundant in the mycobiome of both murine models and human patients with pancreatic tumors. Whether this represents a causal factor or a consequence of tumorigenesis remains unclear, emphasizing the need for further investigation into microbiome–mycobiome interactions [27]. Gut dysbiosis is frequently associated with increased intestinal permeability [28]. Under physiological conditions, bacteria and their products are contained and locally cleared by mucosal immune defenses. However, enhanced permeability allows translocation of bacterial components such as lipopolysaccharide (LPS)—a Gram-negative cell wall constituent that activates Toll-like receptor 4 (TLR4)—thereby inducing cytokine release, oxidative stress, and inflammation [28]. LPS has also been shown to promote tumor PD-L1 expression through activation of the NF-κB/AKT/MyD88 axis, enhancing resistance to T-cell-mediated cytotoxicity both in vitro and in vivo [29]. Microbial metabolites further modulate pancreatic oncogenesis through diverse mechanisms. Short-chain fatty acids (SCFAs)—including acetate, propionate, butyrate, and lactate—produced by bacterial fermentation of non-digestible carbohydrates, exert protective effects [30]. Acetate mitigates pancreatitis, thereby reducing a significant risk factor for tumorigenesis, while butyrate exhibits cytostatic properties in vitro by suppressing the proliferation of pancreatic adenocarcinoma cells [30]. Conversely, secondary bile acids display pro-tumorigenic activity. Microbial dehydroxylation and deconjugation of primary bile acids generate lithocholic acid (LCA), deoxycholic acid (DCA), and ursodeoxycholic acid (UDCA), a process mediated by Bacteroides, Firmicutes, and Lactobacillus species through their enzymatic repertoire [31,32]. Elevated levels of unconjugated bile acids have been reported in patients with pancreatic adenocarcinoma by O’Rees et al. [33]. Mechanistically, DCA binds the Takeda G protein-coupled receptor 5 (TGR5), activating the EGFR/STAT3 signaling pathway and promoting cell cycle progression [31]. In contrast, trimethylamine N-oxide (TMAO) has been associated with a reduced risk of pancreatic carcinogenesis. By activating the type I interferon (IFN) pathway, TMAO exerts anti-inflammatory effects and has been linked to improved survival in pancreatic cancer patients [34]. Another signaling pathway implicated in pancreatic oncogenesis is the mammalian target of rapamycin (mTOR) cascade, which regulates autophagy, cell growth, and cytoskeletal dynamics [35]. Collectively, these findings underscore the intricate interplay between the gut microbiota, microbial metabolites, and host signaling pathways in pancreatic carcinogenesis. They highlight the need for further mechanistic studies to clarify causality and to support the development of microbiome-targeted therapeutic strategies.

3.3. Intratumoral Microbiota in Pathogenesis and Progression of PDAC

Historically considered a sterile organ, the pancreas is now recognized to hold its own distinct microbiota [36]. Microbial species originating from the oral cavity, stomach, and intestine may colonize the common bile duct and subsequently migrate into the pancreatic duct [37]. The precise mechanisms by which microorganisms reach pancreatic tissue remain a matter of debate. One proposed pathway involves the translocation of duodenal microbes through the pancreatic ductal system, while alternative hypotheses suggest dissemination via mesenteric venous drainage or the lymphatic network [38,39]. Emerging evidence suggests that the intratumoral microbiota may play a role in pancreatic cancer development. Riquelme et al. analyzed microbiota directly isolated from resected pancreatic ductal adenocarcinoma (PDAC) tissue and demonstrated that patients with long-term survival (≥5 years after surgery) exhibited significantly higher alpha-diversity, as determined by 16S rRNA gene sequencing [40]. Similarly, beta-diversity analyses revealed an enrichment of Alphaproteobacteria, Sphingobacteria, and Flavobacteria in long-term survivors, whereas Clostridia and Bacteroides predominated in tumors from short-term survivors [40]. Further studies have suggested that the presence of specific microbial taxa within the tumor microenvironment—such as Enterobacter, Enterococcus, and Escherichia coli (E. coli) bactibilia—as well as chronic infection with hepatitis B or C viruses, may contribute to pancreatic carcinogenesis [41]. Nonetheless, it remains unclear whether these microbial communities act as active participants in tumor initiation and progression or merely represent secondary colonizers of already neoplastic tissue.

4. Gut Microbiota Influence on Chemotherapy and Immunotherapy

The gut microbiota has recently been identified as one of the “enabling characteristics” within the updated framework of the hallmarks of cancer [42]. A growing body of evidence from in vitro studies, animal models, and clinical investigations demonstrates that the microbiota contributes not only to cancer initiation and progression but also to the modulation of responses to chemotherapy and immunotherapy, thereby influencing mechanisms of therapeutic resistance [43,44]. Moreover, emerging data highlight its role in cancer therapy-related toxicities, further underscoring its potential significance as a therapeutic target.

4.1. Impact of the Microbiota on Therapeutic Efficacy in PDAC

Approximately 80% of pancreatic ductal adenocarcinoma (PDAC) cases are diagnosed at advanced stages, precluding surgical resection [45]. Consequently, chemotherapy remains the main treatment for most patients [46]. For unresectable PDAC, first-line regimens include combinations of 5-fluorouracil (5-FU), irinotecan, and oxaliplatin with folinic acid (FOLFIRINOX), or gemcitabine with nab-paclitaxel (GnP) [46,47]. However, PDAC is frequently characterized by both intrinsic and rapidly acquired resistance to chemotherapy [48], with fewer than half of patients exhibiting a favorable response [49]. This pronounced chemoresistance is attributed to several defining features of PDAC, including its complex genetic landscape, metabolic reprogramming, and the unique properties of its tumor microenvironment (TME) [50]. Hallmarks of the PDAC TME, such as a dense desmoplastic stroma composed of extracellular matrix (ECM), collagen, and activated pancreatic stellate cells, create a physical barrier that limits drug penetration, reduces tumor perfusion, and impedes immune cell infiltration [51,52]. Furthermore, the relatively high bacterial load within the PDAC TME compared with other malignancies underlines its susceptibility to microbial colonization [53], highlighting opportunities for microbiome-directed approaches to overcome drug resistance. Several studies have linked alterations in gut microbiota composition and specific microbial taxa to reduced cytotoxic efficacy of chemotherapy [43,44]. Geller et al. demonstrated that Mycoplasma hyorhinis within human dermal fibroblasts induced gemcitabine resistance in both in vitro and murine models [54]. Similarly, members of the Gammaproteobacteria can inactivate gemcitabine through the expression of a long isoform of cytidine deaminase (CDD), which converts the drug into its inactive metabolite [54]. In a cohort of 113 PDAC tissue samples, Gammaproteobacteria were detected in 76% of cases. In colorectal cancer murine models, the introduction of gemcitabine-degrading bacteria conferred resistance, which was reversed by co-administration of ciprofloxacin [54]. Consistent with these findings, a retrospective analysis by Weniger et al. reported that patients with PDAC receiving adjuvant gemcitabine had improved progression-free survival (PFS) in the absence of Klebsiella pneumoniae in bile cultures compared with those harboring the bacterium [55]. Additional evidence suggests that Escherichia coli (E. coli) contributes to chemoresistance. Lehouritis et al. demonstrated that a non-pathogenic E. coli strain altered the chemical structure of several chemotherapeutic agents, including gemcitabine, thereby inducing resistance in both in vitro and in vivo murine models [56]. In colorectal and pancreatic models, Fusobacterium nucleatum has been associated with 5-FU resistance through inhibition of apoptotic pathways via a TLR4/MYD88-dependent mechanism [57,58,59]. Conversely, certain commensal bacteria may enhance chemotherapy efficacy. Non-enterotoxigenic Bacteroides fragilis has been shown to stimulate T-cell-mediated immune responses that potentiate oxaliplatin-induced tumor apoptosis within the TME [60]. Likewise, butyrate-producing bacteria have been associated with improved oxaliplatin responsiveness through activation of CD8+ T cells, while antibiotic-mediated depletion of these species increased tumor resistance [60]. Panebianco et al. further demonstrated that butyrate enhanced the efficacy of gemcitabine in in vitro PDAC models, primarily by inducing apoptosis [61]. Beyond direct bacterial mechanisms, microbial metabolites also modulate the drug response [43,44]. Indole-3-acetic acid (3-IAA), a tryptophan-derived metabolite, was found at higher concentrations in patients who responded to chemotherapy [62]. This compound promotes the accumulation of reactive oxygen species (ROS), compromises the metabolic integrity of cancer cells, and enhances treatment efficacy. Strategies such as fecal microbiota transplantation, dietary interventions to increase tryptophan intake, and oral 3-IAA supplementation have been shown to improve chemotherapy outcomes in humanized gnotobiotic PDAC mouse models [62]. In addition to chemotherapy resistance, the gut microbiota has been implicated in modulating immunotherapy responses in PDAC. Although immune checkpoint inhibitors (ICIs) have demonstrated clinical benefit in several malignancies, fewer than 1% of PDAC patients respond [63]. This limited efficacy is attributed mainly to the immunosuppressive TME and the low tumor mutational burden characteristic of PDAC [64]. Significantly, microbiota-driven modulation of the TME may influence ICI responsiveness. Pushalkar et al. showed that antibiotic-mediated microbial ablation upregulated PD-1 expression and enhanced the antitumor effects of PD-1 blockade in a PDAC mouse model [38].
Collectively, these findings suggest that microbial communities and their metabolites play a role in both chemotherapy resistance and immune evasion in PDAC. Microbiota-targeted interventions, including dietary modification, prebiotics, probiotics, selective antibiotics, and fecal microbiota transplantation, represent promising strategies to enhance therapeutic efficacy (Table 1).

4.2. Impact of the Microbiota on Antitumor Therapy-Related Toxicities

As previously discussed, the gut microbiota plays a pivotal role in modulating the metabolism of chemotherapeutic drugs, thereby influencing both treatment efficacy and the host’s susceptibility to therapy-related toxicities [65]. Preclinical studies have shown that 5-fluorouracil (5-FU) increases the relative abundance of pathogenic taxa such as Escherichia, Enterococcus spp., and Clostridium, predisposing patients to mucositis, bacteremia, and sepsis [66,67]. Consistent with these findings, cisplatin-induced toxicity has been attenuated by the administration of D-methionine, which supports the growth of beneficial taxa, including Lachnospiraceae and Lactobacillus [66,67]. The gut microbiota also modulates irinotecan-induced toxicity. Irinotecan is converted into its active metabolite, SN-38, through hepatic glucuronidation and subsequently excreted into the intestinal lumen, where microbial β-glucuronidases hydrolyze it back to SN-38. This reactivated metabolite causes direct mucosal injury, leading to severe diarrhea that frequently requires dose reduction or treatment discontinuation [68,69,70]. Dysbiosis—whether induced by chemotherapy or by the disease itself—results in the overrepresentation of β-glucuronidase-rich taxa such as Proteobacteria, Clostridium clusters, and Fusobacteria, further exacerbating gastrointestinal toxicity and inflammation [71]. Given its pivotal role in chemotherapy-associated toxicities, modulation of the gut microbiota has emerged as a promising therapeutic strategy. Preclinical models have demonstrated that selective inhibition of microbial β-glucuronidases, combined with probiotic supplementation, markedly reduces SN-38 reactivation, thereby limiting mucosal injury, oxidative stress, and inflammatory cytokine release [71]. Panebianco et al. further reported that in vivo probiotic administration counteracted dysbiosis induced by gemcitabine plus nab-paclitaxel, decreased chemotherapy-related side effects and stromal activation, improved microbial diversity, and increased the abundance of SCFA-producing bacteria [72]. Despite these encouraging preclinical findings, rigorously designed randomized clinical trials are still required to confirm the safety and efficacy of microbiota-targeted interventions for mitigating treatment-related toxicities in pancreatic cancer (Table 2).

5. Microbiota Modulation in the Treatment of PDAC: Classic and New Therapeutic Strategies

The intestinal microbiota has emerged as a critical component in PDAC, shaping tumorigenesis, antitumor immunity, and therapeutic responsiveness. Multiple therapeutic strategies, including antibiotics, probiotics, dietary interventions, engineered bacteria, oncolytic viruses, and fecal microbiota transplantation (FMT), are currently under investigation to restore microbial homeostasis and improve treatment outcomes [21]. Increasingly, modulation of the microbiota is a key determinant of therapeutic efficacy in both chemotherapy and immunotherapy. In the context of immunotherapy, particularly with immune checkpoint inhibitors (ICIs) targeting PD-1/PD-L1 and CTLA-4, the gut microbiota modulates immune responses. It can regulate both systemic and intratumoral immunity by affecting the expression of checkpoint proteins and promoting remodeling of the tumor microenvironment. Through these mechanisms, the microbiota may enhance the efficacy of checkpoint blockade and improve response rates [73]. In chemotherapy, regimens like gemcitabine and FOLFIRINOX are challenged by desmoplastic stroma and multidrug resistance. Microbial communities can affect drug metabolism, bioactivation, and tissue penetration [74]. Targeted manipulation of the gut, oral, or intratumoral microbiota is a promising strategy for overcoming therapeutic resistance and enhancing the effects of established cytotoxic and immune-based treatments. This approach could open new avenues to improve clinical outcomes in patients with pancreatic ductal adenocarcinoma (PDAC).

5.1. Antibiotics

Antibiotic therapy has shown promising results in altering the microbiota and improving the response to chemotherapy, particularly gemcitabine. In preclinical models, administration of broad-spectrum antibiotics reduced tumor burden by depleting microbial populations [75]. Two independent studies further demonstrated that antibiotic-mediated microbiota ablation reprograms antitumor immune responses and influences PDAC progression. Sethi et al. reported that antibiotics delayed tumor growth in subcutaneous PDAC models by promoting the infiltration of IFN-γ-producing cytotoxic T cells, while suppressing immunosuppressive T cells that secrete IL-17A and IL-10 [76]. Similarly, Pushalkar et al. observed that microbial ablation in orthotopic PDAC models decreased tumor burden and induced an immunostimulatory microenvironment, characterized by increased M1 macrophages, CD4+ Th1 cells, and CD8+ T cells, alongside a reduction in MDSCs and TLR2/5 signaling [38]. This immune reprogramming may also enhance chemotherapy efficacy, as intratumoral Gammaproteobacteria can express the long isoform of cytidine deaminase (CDD), which inactivates gemcitabine and promotes resistance [54,77]. Elevated intratumoral bacterial loads have been correlated with reduced gemcitabine efficacy, an effect reversed following postoperative antibiotic therapy [78,79]. Clinical data corroborate these preclinical findings. Patients receiving antibiotics during gemcitabine–nab-paclitaxel (GnP) treatment exhibited longer progression-free survival (5.8 vs. 2.7 months) and improved overall outcomes [80]. Retrospective analyses further support the benefit of concurrent antibiotic therapy, demonstrating improved overall survival (OS) and progression-free survival (PFS) when combined with gemcitabine, but not with fluoropyrimidines, suggesting a drug-specific interaction with the microbiota [81]. In a cohort of 216 patients, first-line gemcitabine plus antibiotics significantly prolonged both OS and PFS, whereas fluoropyrimidine-based regimens yielded only marginal PFS benefits [80]. Among antibiotic classes, specific agents appear more effective. Ciprofloxacin has been shown to counteract gemcitabine resistance mediated by Gammaproteobacteria [54]. Both macrolides and quinolones, when used in combination with chemotherapy, are associated with survival gains of approximately 2–3 months in metastatic PDAC [80]. Quinolones are particularly effective in patients colonized with Klebsiella pneumoniae [55], while macrolide therapy lasting more than three days has been independently correlated with improved OS and PFS [80]. Despite these encouraging findings, concerns remain regarding prolonged use of broad-spectrum antibiotics. Documented risks include gastrointestinal, hepatic, and hematologic toxicities [82]; dysbiosis and opportunistic infections [83]; and the emergence of antimicrobial resistance. To mitigate systemic toxicity, localized antibiotic delivery systems have been explored. In a murine colon carcinoma model, microdevices implanted with ampicillin and chloramphenicol directly into tumors inhibited growth and enhanced apoptosis. When combined with gemcitabine, these effects were further amplified, underscoring the critical role of intratumoral bacteria in mediating chemoresistance [54] (Table 3).

5.2. Probiotics and Prebiotics

Probiotics have gained increasing attention as non-invasive strategies to modulate microbial communities. Because microbes from the oral–gut axis may translocate to the pancreas via the pancreatic duct, oral probiotic administration represents a plausible route of intervention, although clinical validation remains necessary [84]. Lactobacillus species—including L. paracasei, L. reuteri, and L. rhamnosus—exhibit antitumor activity by inhibiting tumor growth and metastasis while enhancing the efficacy of gemcitabine. These effects involve p53 activation, cell cycle arrest, and modulation of epithelial–mesenchymal transition (EMT) markers [83,85,86]. In murine models, co-administration of Lactobacillus spp with gemcitabine significantly attenuated PanIN progression and improved hepatic enzyme profiles, suggesting both enhanced tolerability and therapeutic efficacy [86]. Additional studies have shown that L. reuteri suppresses PDAC progression by promoting natural killer (NK) cell infiltration, and higher fecal abundance of Lactobacillus correlates with improved survival [87]. Beyond probiotics, postbiotic compounds derived from these microorganisms also display therapeutic potential. Ferrichrome, secreted by L. casei, exerts antineoplastic effects even in 5-FU-resistant cell lines via p53-mediated apoptotic pathways [88]. Similarly, Bifidobacterium longum attenuates PDAC progression by enhancing CD8+ T-cell infiltration and reducing immunosuppressive cell subsets [89]. This species also synergizes with anti-PD-L1 therapy through dendritic cell activation [90]. Notably, when combined with vancomycin and neomycin, B. longum further improved antitumor responses and promoted CD8+ T-cell recruitment in murine models [91]. Another relevant species, Akkermansia muciniphila, has been shown to enhance CD8+ T-cell-mediated immunity and improve responses to immune checkpoint inhibitors [92,93]. Further evidence supports the efficacy of synbiotics—combinations of probiotics and prebiotics—in optimizing host–microbe interactions. In a randomized trial involving 90 PDAC patients, perioperative administration of probiotics plus inulin increased intratumoral CD8+ T-cell infiltration and IFN-γ expression, as demonstrated by immunohistochemistry of resected tumors [91]. Synbiotic therapy also reduced circulating levels of pro-inflammatory cytokines (IL-1β, IL-6, and IL-10) [91]. Clinically, these effects were associated with fewer postoperative complications and a lower incidence of bacteremia, underscoring the potential role of synbiotics in perioperative management [94]. Postbiotics—defined as bioactive microbial-derived metabolites—represent another promising frontier in microbiota-based therapy. SCFA, particularly butyrate, together with metabolites such as indole-3-acetic acid, exert regulatory effects on both tumor and immune cells. Butyrate, produced through microbial fermentation of dietary fibers, enhances gemcitabine efficacy by inducing oxidative stress and autophagy [62]. It also reduces tumor proliferation by inhibiting histone deacetylase (HDAC) and modulating epigenetics, highlighting its multifaceted role in pancreatic cancer therapy [95] (Table 4).

5.3. Engineered Bacteria

The potential of engineered bacteria as ‘living drugs’ to directly target the tumor microenvironment is a source of great optimism. For instance, hypoxia-responsive Escherichia coli strains have been genetically modified to secrete cyst(e)inase (CGL), leading to the depletion of intracellular cysteine and cystine pools. This process induces ferroptosis through lipid peroxidation while also activating antitumor immune responses [96]. Other approaches involve the use of synthetic Salmonella strains engineered to deliver STING agonists via outer membrane vesicles (OMVs). These bacterial strains enhance immune cell infiltration and act synergistically with immune checkpoint inhibitors [97]. Similarly, attenuated Salmonella Typhimurium strains engineered to secrete immunomodulatory proteins have been shown to inhibit PDAC growth and modulate cytokine profiles [98]. Another innovative strategy involves the functionalization of Lactobacillus rhamnosus GG with a gallium–polyphenol network (LGG@Ga-poly). This formulation effectively depletes Proteobacteria and lipopolysaccharides (LPSs), remodels the tumor immune microenvironment, and enhances the efficacy of immunotherapy [99] (Table 5).

5.4. Non-Bacterial Microbiome Components: Bacteriophages and Oncolytic Viruses

Lytic bacteriophages provide a highly selective method for eliminating pathogenic bacteria associated with tumors. Additionally, phage display technologies have enabled the identification of tumor-specific peptides, such as MCA1, which bind to PDAC cells with high specificity and nanomolar affinity. This presents potential applications for imaging and targeted drug delivery [100]. Fusion proteins derived from bacteriophages have also been employed to functionalize liposomes containing doxorubicin, thereby enhancing tumor selectivity and cytotoxic potency [101]. Similarly, polymeric micelles functionalized with phage-derived proteins and loaded with paclitaxel have demonstrated improved tumor targeting, increased drug solubility, and reduced off-target toxicity [102]. Oncolytic viruses (OVs) represent a distinct class of biotherapeutics capable of interacting with the microbiota and remodeling the tumor microenvironment. Adenoviruses have been extensively investigated due to their stability and high transduction efficiency. While first-generation vectors such as ONYX-015 exhibited limited replication capacity and were rapidly neutralized by host immunity [103,104], next-generation adenoviral platforms have been engineered to overcome antiviral resistance [105], enhance tumor radiosensitivity [106], and modulate stromal components [107]. Herpes simplex virus (HSV)-based OVs have been shown to induce apoptosis and necrosis of tumor cells in preclinical PDAC models while synergizing with gemcitabine [108]. Naturally mutated HSV strains have further demonstrated the ability to activate CD4+/CD8+ T cells and NK cells in early-phase clinical trials [109]. Vaccinia virus strains have also exhibited antitumor effects, either as monotherapy or in combination with chemotherapy and radiotherapy, with encouraging preclinical outcomes and ongoing clinical evaluation [110,111,112]. Reoviruses, which selectively replicate in RAS-mutant cells, are particularly well suited for PDAC. Pelareorep, a clinically tested reovirus, has shown limited efficacy as a single agent, but ongoing studies are exploring its synergistic potential with immune checkpoint inhibitors [113]. Similarly, the Coxsackievirus B3 strain PD-H has demonstrated potent lytic activity in vitro and significant tumor suppression in vivo, although its optimal efficacy may depend on combination strategies targeting the stromal compartment [114] (Table 6).

5.5. Fecal Microbiota Transplantation

Fecal microbiota transplantation (FMT) is rapidly emerging as a promising strategy to modulate both intestinal and tumor-associated microbiota in PDAC. Preclinical studies consistently demonstrate that FMT derived from immunotherapy responders or long-term PDAC survivors reduces tumor burden and enhances T-cell infiltration [75]. Riquelme et al. reported that long-term survivors exhibited a more diverse intratumoral microbiota, including Pseudoxanthomonas, Streptomyces, Saccharopolyspora, and Bacillus clausii (B. clausii), which correlated with improved clinical outcomes [40]. Similarly, Tintelnot et al. found that FMT from chemotherapy responders increased therapeutic sensitivity in murine models, whereas FMT from healthy donors inhibited tumor progression [62]. Growing evidence supports FMT as a valuable adjunct to immunotherapy [115]. By reshaping the gut microbiota, FMT has been shown to restore responsiveness to immune checkpoint inhibitors (ICIs) and to reduce the incidence and severity of immune-related adverse events (irAEs). Although PDAC-specific clinical trials are still lacking, studies in other malignancies provide proof of principle. In melanoma, FMT from responder patients reversed resistance to anti-PD-1/PD-1/PD-L1 therapy, and similar effects were observed using healthy donor-derived material [116,117,118]. In one clinical trial, three of fifteen anti-PD-1-refractory melanoma patients responded to FMT combined with pembrolizumab [119]. In another Phase I trial, FMT followed by anti-PD-1 reinduction achieved one complete and two partial responses among ten patients [116]. Nevertheless, safety concerns persist. A notable adverse event involved transmission of an ESBL-producing Escherichia coli strain from a shared donor, resulting in bacteremia and one fatality [120]. Despite this, FMT has shown clinical benefit in managing ICI-induced colitis. Several reports document its efficacy in steroid- and biologic-refractory cases, allowing continuation of cancer immunotherapy and potentially improving adherence to treatment protocols [121,122,123,124]. FMT may also mitigate radiotherapy-associated toxicity. In patients with refractory radiation enteritis, FMT led to clinical improvement without serious adverse events [125]. In a larger cohort, it was associated with improved gastrointestinal quality of life and nutritional status over a six-month follow-up [126]. In summary, although PDAC-specific trials remain limited, the rationale for microbiota-based interventions is compelling. Integration of FMT with chemotherapy, radiotherapy, or immunotherapy may enhance efficacy while reducing toxicity, particularly in neoadjuvant and perioperative settings. Further clinical studies are urgently needed to validate the safety, efficacy, and long-term benefits of this approach in PDAC management. (Table 7)

5.6. Future Direction

Several critical questions remain unresolved and are now the focus of ongoing clinical investigations. These include the long-term effects and safety profile of fecal microbiota transplantation (FMT) in oncology patients; the identification of specific microbial signatures associated with clinical features, molecular subtypes, or prognosis in PDAC; and the extent to which microbiota composition, microbial metabolites, and immune modulation influence disease progression and therapeutic response. To address these questions, multiple clinical trials are currently underway. The Microbiome of Pancreatic Cancer: “PANDEMIC study” (NCT04274972) is prospectively evaluating whether distinct gut microbial patterns correlate with disease characteristics and outcomes. The PDA-MAPS trial (NCT04922515) is conducting an integrated analysis of oral, gut, and tumor-associated microbiota, combined with genomic data, to determine whether microbiome-based subtypes of PDAC can be defined. Metabolomic and Immune–Microbiome Profiling for Unresectable Pancreatic Cancer (NCT07036978) aims to elucidate how microbial metabolites and host immune responses modulate tumor behavior in advanced disease. In addition, an early-phase study, “Fecal Microbial Transplants for the Treatment of Pancreatic Cancer” (NCT04975217), is assessing the safety and immunomodulatory potential of FMT in patients undergoing preoperative management. In contrast, issues related to the standardization, scalability, and biosafety of engineered bacterial- and viral-based therapies remain largely unaddressed by current clinical trials. Collectively, these efforts underscore a growing recognition that the microbiome may provide critical insights—and potentially novel therapeutic avenues—for improving outcomes in pancreatic cancer.

6. Conclusions

The microbiota is increasingly recognized not only as a driver of PDAC pathogenesis but also as a potent modulator of therapeutic response. From the oral axis to the tumor microenvironment, distinct microbial taxa and their metabolites can either suppress cytotoxic efficacy or, conversely, enhance immune surveillance. Preclinical studies have demonstrated that antibiotics, probiotics, engineered bacteria, bacteriophages, oncolytic viruses, and fecal microbiota transplantation can remodel the desmoplastic and immunosuppressive tumor niche, restoring sensitivity to both chemotherapy and immunotherapy. These findings delineate a new therapeutic frontier. Microbiota modulation—through antibiotics, probiotics, synbiotics, postbiotics, engineered “living drugs,” bacteriophages, oncolytic viruses, or fecal transplantation—holds the potential to overcome chemoresistance, restore immunogenicity, and mitigate treatment-related toxicity. Early clinical evidence, particularly with synbiotic therapy and responder-derived FMT, indicates promising translational potential. However, rigorous clinical validation remains essential. Well-designed, adequately powered trials are necessary to define the efficacy, safety, and optimal integration of microbiota-based approaches into existing treatment regimens. Ultimately, harnessing the microbiome may help transform one of oncology’s most treatment-refractory malignancies into a disease amenable to precision immunotherapy.

Author Contributions

Conceptualization, M.C. and E.C.N.; methodology, F.F.; validation, F.F., A.G.; data curation, G.P.; writing—original draft preparation, F.D. and E.C.; writing—review and editing, S.S.D.L. and S.A.; visualization, E.C.N.; supervision, M.C. and G.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
PDACPancreatic ductal adenocarcinoma
MeSHMedical subject headings
SNPSingle-nucleotide polymorphism
LPSLipopolysaccharide
TLR4Toll-like receptor 4
MyD88Myeloid differentiation primary response 88
PD-L1Programmed death-ligand 1
NF-κBNuclear factor kappa-light-chain-enhancer of activated B cells
AKTProtein kinase B
SCFAsShort-chain fatty acids
LCALithocholic acid
DCADeoxycholic acid
UDCAUrsodeoxycholic acid
TGR5Takeda G protein-coupled receptor 5
EGFREpidermal growth factor receptor
STAT3Signal transducer and activator of transcription 3
TMAOTrimethylamine N-oxide
IFNInterferon
mTORMammalian target of rapamycin
ECMExtracellular matrix
TMETumor microenvironment
5-FU5-Fluorouracil
GnPGemcitabine plus nab-paclitaxel
PFSProgression-free survival
OSOverall survival
CDDCytidine deaminase
TLR2Toll-like receptor 2
TLR5Toll-like receptor 5
MDSCsMyeloid-derived suppressor cells
ROSReactive oxygen species
3-IAAIndole-3-acetic acid
ICI/ICIsImmune checkpoint inhibitor(s)
PD-1Programmed cell death protein 1
CD8+Cytotoxic T lymphocyte (CD8 positive)
CD4+Helper T lymphocyte (CD4 positive)
Th1T helper 1 cell
M1M1 macrophage (pro-inflammatory phenotype)
IL-1βInterleukin 1 beta
IL-6Interleukin 6
IL-10Interleukin 10
IL-17AInterleukin 17A
FOLFIRINOX5-FU, leucovorin, irinotecan, oxaliplatin
GUSβ-glucuronidase
SN-38/SN-38G7-ethyl-10-hydroxycamptothecin/SN-38 glucuronide
EMTEpithelial–mesenchymal transition
NKNatural killer cell
FMTFecal microbiota transplantation
STINGStimulator of interferon genes
OMV/OMVsOuter membrane vesicle(s)
OV/OVsOncolytic virus(es)
HSVHerpes simplex virus
ONYX-015First-generation oncolytic adenovirus vector
LGGLactobacillus rhamnosus GG
LGG@Ga-polyLGG functionalized with gallium–polyphenol network
CGLCyst(e)inase enzyme
RASRat sarcoma viral oncogene family
KRAS^G12DKRAS gene mutation (glycine 12 to aspartate)
PanINPancreatic intraepithelial neoplasia
KPCKras^G12D; Trp53^R172H; Pdx1-Cre mouse model
ATCCAmerican Type Culture Collection
MCA1Phage-derived tumor-targeting peptide
irAEsImmune-related adverse events
GIGastrointestinal

References

  1. Afzaal, M.; Saeed, F.; Shah, Y.A.; Hussain, M.; Rabail, R.; Socol, C.T.; Hassoun, A.; Pateiro, M.; Lorenzo, J.M.; Rusu, A.V.; et al. Human gut microbiota in health and disease: Unveiling the relationship. Front. Microbiol. 2022, 13, 999001. [Google Scholar] [CrossRef] [PubMed]
  2. Pickard, J.M.; Zeng, M.Y.; Caruso, R.; Núñez, G. Gut microbiota: Role in pathogen colonization, immune responses, and inflammatory disease. Immunol. Rev. 2017, 279, 70–89. [Google Scholar] [CrossRef] [PubMed]
  3. Mishra, S.P.; Wang, B.; Jain, S.; Ding, J.; Rejeski, J.; Furdui, C.M.; Kitzman, D.W.; Taraphder, S.; Brechot, C.; Kumar, A.; et al. A mechanism by which gut microbiota elevates permeability and inflammation in obese/diabetic mice and human gut. Gut 2023, 72, 1848–1865. [Google Scholar] [CrossRef] [PubMed]
  4. Flandroy, L.; Poutahidis, T.; Berg, G.; Clarke, G.; Dao, M.-C.; Decaestecker, E.; Furman, E.; Haahtela, T.; Massart, S.; Plovier, H.; et al. The impact of human activities and lifestyles on the interlinked microbiota and health of humans and of ecosystems. Sci. Total. Environ. 2018, 627, 1018–1038. [Google Scholar] [CrossRef]
  5. Niccolai, E.; Boem, F.; Emmi, G.; Amedei, A. The link “Cancer and autoimmune diseases” in the light of microbiota: Evidence of a potential culprit. Immunol. Lett. 2020, 222, 12–28. [Google Scholar] [CrossRef]
  6. Aburto, M.R.; Cryan, J.F. Gastrointestinal and brain barriers: Unlocking gates of communication across the microbiota–gut–brain axis. Nat. Rev. Gastroenterol. Hepatol. 2024, 21, 222–247. [Google Scholar] [CrossRef]
  7. Signoretti, M.; Roggiolani, R.; Stornello, C.; Fave, G.D.; Capurso, G. Gut microbiota and pancreatic diseases. Minerva Gastroenterol. 2017, 63, 399–410. [Google Scholar] [CrossRef]
  8. Ren, S.; Qian, L.-C.; Cao, Y.-Y.; Daniels, M.J.; Song, L.-N.; Tian, Y.; Wang, Z.-Q. Computed tomography-based radiomics diagnostic approach for differential diagnosis between early- and late-stage pancreatic ductal adenocarcinoma. World J. Gastrointest. Oncol. 2024, 16, 1256–1267. [Google Scholar] [CrossRef]
  9. Leiphrakpam, P.D.; Chowdhury, S.; Zhang, M.; Bajaj, V.; Dhir, M.; Are, C. Trends in the Global Incidence of Pancreatic Cancer and a Brief Review of its Histologic and Molecular Subtypes. J. Gastrointest. Cancer 2025, 56, 1–13. [Google Scholar] [CrossRef]
  10. Li, J.J.; Zhu, M.; Kashyap, P.C.; Chia, N.; Tran, N.H.; McWilliams, R.R.; Bekaii-Saab, T.S.; Ma, W.W. The role of microbiome in pancreatic cancer. Cancer Metastasis Rev. 2021, 40, 777–789. [Google Scholar] [CrossRef]
  11. Routy, B.; Gopalakrishnan, V.; Daillère, R.; Zitvogel, L.; Wargo, J.A.; Kroemer, G. The gut microbiota influences anticancer immunosurveillance and general health. Nat. Rev. Clin. Oncol. 2018, 15, 382–396. [Google Scholar] [CrossRef]
  12. Sfanos, K.S. Intratumoral Bacteria as Mediators of Cancer Immunotherapy Response. Cancer Res. 2023, 83, 2985–2986. [Google Scholar] [CrossRef]
  13. Baker, J.L.; Welch, J.L.M.; Kauffman, K.M.; McLean, J.S.; He, X. The oral microbiome: Diversity, biogeography and human health. Nat. Rev. Microbiol. 2023, 22, 89–104. [Google Scholar] [CrossRef]
  14. Fan, X.; Alekseyenko, A.V.; Wu, J.; Peters, B.A.; Jacobs, E.J.; Gapstur, S.M.; Purdue, M.P.; Abnet, C.C.; 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, 120–127. [Google Scholar] [CrossRef]
  15. Slocum, C.; Kramer, C.; Genco, C.A. Immune dysregulation mediated by the oral microbiome: Potential link to chronic inflammation and atherosclerosis. J. Intern. Med. 2016, 280, 114–128. [Google Scholar] [CrossRef] [PubMed]
  16. Maisonneuve, P.; Amar, S.; Lowenfels, A.B. Periodontal disease, edentulism, and pancreatic cancer: A meta-analysis. Ann. Oncol. 2017, 28, 985–995. [Google Scholar] [CrossRef] [PubMed]
  17. Michaud, D.S.; Fu, Z.; Shi, J.; Chung, M. Periodontal Disease, Tooth Loss, and Cancer Risk. Epidemiol. Rev. 2017, 39, 49–58. [Google Scholar] [CrossRef] [PubMed]
  18. Hujoel, P.P.; Drangsholt, M.; Spiekerman, C.; Weiss, N.S. An Exploration of the Periodontitis–Cancer Association. Ann. Epidemiology 2003, 13, 312–316. [Google Scholar] [CrossRef]
  19. Michaud, D.S.; Izard, J.; Wilhelm-Benartzi, C.S.; You, D.-H.; Grote, V.A.; Tjonneland, A.; Dahm, C.C.; 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, 1764–1770. [Google Scholar] [CrossRef]
  20. Saba, E.; Farhat, M.; Daoud, A.; Khashan, A.; Forkush, E.; Menahem, N.H.; Makkawi, H.; Pandi, K.; Angabo, S.; Kawasaki, H.; et al. Oral bacteria accelerate pancreatic cancer development in mice. Gut 2024, 73, 770–786. [Google Scholar] [CrossRef]
  21. Guo, X.; Shao, Y. Role of the oral-gut microbiota axis in pancreatic cancer: A new perspective on tumor pathophysiology, diagnosis, and treatment. Mol. Med. 2025, 31, 1–19. [Google Scholar] [CrossRef]
  22. Papa, V.; Schepis, T.; Coppola, G.; Chiappetta, M.F.; Del Vecchio, L.E.; Rozera, T.; Quero, G.; Gasbarrini, A.; Alfieri, S.; Papa, A. The Role of Microbiota in Pancreatic Cancer. Cancers 2023, 15, 3143. [Google Scholar] [CrossRef]
  23. 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, 222–238. [Google Scholar] [CrossRef]
  24. Han, Y.; Cao, B.; Tang, J.; Wang, J. A comprehensive multi-omics analysis uncovers the associations between gut microbiota and pancreatic cancer. Front. Microbiol. 2025, 16, 1592549. [Google Scholar] [CrossRef]
  25. Lee, A.A.; Wang, Q.-L.; Kim, J.; Babic, A.; Zhang, X.; Perez, K.; Ng, K.; Nowak, J.; Rifai, N.; Sesso, H.D.; et al. Helicobacter pylori Seropositivity, ABO Blood Type, and Pancreatic Cancer Risk From 5 Prospective Cohorts. Clin. Transl. Gastroenterol. 2023, 14, e00573. [Google Scholar] [CrossRef]
  26. Sammallahti, H.; Kokkola, A.; Rezasoltani, S.; Ghanbari, R.; Aghdaei, H.A.; Knuutila, S.; Puolakkainen, P.; Sarhadi, V.K. Microbiota Alterations and Their Association with Oncogenomic Changes in Pancreatic Cancer Patients. Int. J. Mol. Sci. 2021, 22, 12978. [Google Scholar] [CrossRef]
  27. Aykut, B.; Pushalkar, S.; Chen, R.; Li, Q.; Abengozar, R.; Kim, J.I.; Shadaloey, S.A.; Wu, D.; Preiss, P.; Verma, N.; et al. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature 2019, 574, 264–267. [Google Scholar] [CrossRef]
  28. Daniel, N.; Farinella, R.; Belluomini, F.; Fajkic, A.; Rizzato, C.; Souček, P.; Campa, D.; Hughes, D.J. The relationship of the microbiome, associated metabolites and the gut barrier with pancreatic cancer. Semin. Cancer Biol. 2025, 112, 43–57. [Google Scholar] [CrossRef]
  29. Yin, H.; Pu, N.; Chen, Q.; Zhang, J.; Zhao, G.; Xu, X.; Wang, D.; Kuang, T.; Jin, D.; Lou, W.; et al. Gut-derived lipopolysaccharide remodels tumoral microenvironment and synergizes with PD-L1 checkpoint blockade via TLR4/MyD88/AKT/NF-κB pathway in pancreatic cancer. Cell Death Dis. 2021, 12, 1–14. [Google Scholar] [CrossRef]
  30. Ratajczak, W.; Rył, A.; Mizerski, A.; Walczakiewicz, K.; Sipak, O.; Laszczyńska, M. Immunomodulatory potential of gut microbiome-derived short-chain fatty acids (SCFAs). Acta Biochim. Pol. 2019, 66, 1–12. [Google Scholar] [CrossRef]
  31. Nagathihalli, N.S.; Beesetty, Y.; Lee, W.; Washington, M.K.; Chen, X.; Lockhart, A.C.; Merchant, N.B. 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, 2062–2072. [Google Scholar] [CrossRef]
  32. Ridlon, J.M.; Kang, D.J.; Hylemon, P.B. Bile salt biotransformations by human intestinal bacteria. J. Lipid Res. 2006, 47, 241–259. [Google Scholar] [CrossRef]
  33. Rees, D.O.; Crick, P.J.; Jenkins, G.J.; Wang, Y.; Griffiths, W.J.; Brown, T.H.; Al-Sarireh, B. Comparison of the composition of bile acids in bile of patients with adenocarcinoma of the pancreas and benign disease. J. Steroid Biochem. Mol. Biol. 2017, 174, 290–295. [Google Scholar] [CrossRef]
  34. Mirji, G.; Worth, A.; Bhat, S.A.; El Sayed, M.; Kannan, T.; Goldman, A.R.; Tang, H.-Y.; Liu, Q.; Auslander, N.; Dang, C.V.; et al. The microbiome-derived metabolite TMAO drives immune activation and boosts responses to immune checkpoint blockade in pancreatic cancer. Sci. Immunol. 2022, 7, eabn0704. [Google Scholar] [CrossRef]
  35. Noureldein, M.H.; Eid, A.A. Gut microbiota and mTOR signaling: Insight on a new pathophysiological interaction. Microb. Pathog. 2018, 118, 98–104. [Google Scholar] [CrossRef]
  36. Lu, W.; Aihaiti, A.; Abudukeranmu, P.; Liu, Y.; Gao, H. Unravelling the role of intratumoral bacteria in digestive system cancers: Current insights and future perspectives. J. Transl. Med. 2024, 22, 545. [Google Scholar] [CrossRef]
  37. Qian, J.; Zhang, X.; Wei, B.; Tang, Z.; Zhang, B. The correlation between gut and intra-tumor microbiota and PDAC: Etiology, diagnostics and therapeutics. Biochim. et Biophys. Acta (BBA)-Rev. Cancer 2023, 1878, 188943. [Google Scholar] [CrossRef]
  38. Pushalkar, S.; Hundeyin, M.; Daley, D.; Zambirinis, C.P.; Kurz, E.; Mishra, A.; Mohan, N.; Aykut, B.; Usyk, M.; Torres, L.E.; et al. The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression. Cancer Discov. 2018, 8, 403–416. [Google Scholar] [CrossRef]
  39. Diehl, G.E.; Longman, R.S.; Zhang, J.-X.; Breart, B.; Galan, C.; Cuesta, A.; Schwab, S.R.; Littman, D.R. Microbiota restricts trafficking of bacteria to mesenteric lymph nodes by CX3CR1hi cells. Nature 2013, 494, 116–120. [Google Scholar] [CrossRef]
  40. Riquelme, E.; Zhang, Y.; Zhang, L.; Montiel, M.; Zoltan, M.; Dong, W.; Quesada, P.; Sahin, I.; Chandra, V.; Lucas, A.S.; et al. Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes. Cell 2019, 178, 795–806.e12. [Google Scholar] [CrossRef]
  41. Kiss, B.; Mikó, E.; Sebő, É.; Toth, J.; Ujlaki, G.; Szabó, J.; Uray, K.; Bai, P.; Árkosy, P. Oncobiosis and Microbial Metabolite Signaling in Pancreatic Adenocarcinoma. Cancers 2020, 12, 1068. [Google Scholar] [CrossRef]
  42. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef] [PubMed]
  43. Beatty, G.L.; Werba, G.; Lyssiotis, C.A.; Simeone, D.M. The biological underpinnings of therapeutic resistance in pancreatic cancer. Genes Dev. 2021, 35, 940–962. [Google Scholar] [CrossRef]
  44. Chrysostomou, D.; Roberts, L.A.; Marchesi, J.R.; Kinross, J.M. Gut Microbiota Modulation of Efficacy and Toxicity of Cancer Chemotherapy and Immunotherapy. Gastroenterology 2022, 164, 198–213. [Google Scholar] [CrossRef]
  45. Giovannetti, E.; van der Borden, C.; Frampton, A.; Ali, A.; Firuzi, O.; Peters, G. Never let it go: Stopping key mechanisms underlying metastasis to fight pancreatic cancer. Semin. Cancer Biol. 2017, 44, 43–59. [Google Scholar] [CrossRef]
  46. Conroy, T.; Hammel, P.; Hebbar, M.; Ben Abdelghani, M.; Wei, A.C.; Raoul, J.-L.; Choné, L.; Francois, E.; Artru, P.; Biagi, J.J.; et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N. Engl. J. Med. 2018, 379, 2395–2406. [Google Scholar] [CrossRef]
  47. Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased Survival in Pancreatic Cancer with nab-Paclitaxel plus Gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef]
  48. Zeng, S.; Pöttler, M.; Lan, B.; Grützmann, R.; Pilarsky, C.; Yang, H. Chemoresistance in Pancreatic Cancer. Int. J. Mol. Sci. 2019, 20, 4504. [Google Scholar] [CrossRef]
  49. Raghavan, S.; Winter, P.S.; Navia, A.W.; Williams, H.L.; DenAdel, A.; Lowder, K.E.; Galvez-Reyes, J.; Kalekar, R.L.; Mulugeta, N.; Kapner, K.S.; et al. Microenvironment drives cell state, plasticity, and drug response in pancreatic cancer. Cell 2021, 184, 6119–6137.e26. [Google Scholar] [CrossRef]
  50. Capula, M.; Perán, M.; Xu, G.; Donati, V.; Yee, D.; Gregori, A.; Assaraf, Y.G.; Giovannetti, E.; Deng, D. Role of drug catabolism, modulation of oncogenic signaling and tumor microenvironment in microbe-mediated pancreatic cancer chemoresistance. Drug Resist. Updat. 2022, 64, 100864. [Google Scholar] [CrossRef]
  51. Hernández-Camarero, P.; López-Ruiz, E.; Marchal, J.A.; Perán, M. Cancer: A mirrored room between tumor bulk and tumor microenvironment. J. Exp. Clin. Cancer Res. 2021, 40, 1–12. [Google Scholar] [CrossRef]
  52. Apte, M.V.; Wilson, J.S.; Lugea, A.; Pandol, S.J. A Starring Role for Stellate Cells in the Pancreatic Cancer Microenvironment. Gastroenterology 2013, 144, 1210–1219. [Google Scholar] [CrossRef]
  53. Dickson, I. Microbiome promotes pancreatic cancer. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 328. [Google Scholar] [CrossRef]
  54. Geller, L.T.; Barzily-Rokni, M.; Danino, T.; Jonas, O.H.; Shental, N.; Nejman, D.; Gavert, N.; Zwang, Y.; Cooper, Z.A.; Shee, K.; et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 2017, 357, 1156–1160. [Google Scholar] [CrossRef]
  55. Weniger, M.; Hank, T.; Qadan, M.; Ciprani, D.; Michelakos, T.; Niess, H.; Heiliger, C.; Ilmer, M.; D’Haese, J.G.; Ferrone, C.R.; et al. Influence of Klebsiella pneumoniae and quinolone treatment on prognosis in patients with pancreatic cancer. Br. J. Surg. 2020, 108, 709–716. [Google Scholar] [CrossRef]
  56. Lehouritis, P.; Cummins, J.; Stanton, M.; Murphy, C.T.; McCarthy, F.O.; Reid, G.; Urbaniak, C.; Byrne, W.L.; Tangney, M. Local bacteria affect the efficacy of chemotherapeutic drugs. Sci. Rep. 2015, 5, 14554. [Google Scholar] [CrossRef]
  57. Ye, C.; Liu, X.; Liu, Z.; Pan, C.; Zhang, X.; Zhao, Z.; Sun, H. Fusobacterium nucleatum in tumors: From tumorigenesis to tumor metastasis and tumor resistance. Cancer Biol. Ther. 2024, 25, 2306676. [Google Scholar] [CrossRef]
  58. Udayasuryan, B.; Ahmad, R.N.; Nguyen, T.T.D.; Umaña, A.; Roberts, L.M.; Sobol, P.; Jones, S.D.; Munson, J.M.; Slade, D.J.; Verbridge, S.S. Fusobacterium nucleatum induces proliferation and migration in pancreatic cancer cells through host autocrine and paracrine signaling. Sci. Signal. 2022, 15, eabn4948. [Google Scholar] [CrossRef]
  59. Yu, T.; 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, 548–563.e16. [Google Scholar] [CrossRef]
  60. Bai, L.; Yan, X.; Lv, J.; Qi, P.; Song, X.; Zhang, L. Intestinal Flora in Chemotherapy Resistance of Biliary Pancreatic Cancer. Biology 2023, 12, 1151. [Google Scholar] [CrossRef]
  61. Panebianco, C.; Villani, A.; Pisati, F.; Orsenigo, F.; Ulaszewska, M.; Latiano, T.P.; 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. [Google Scholar] [CrossRef]
  62. Tintelnot, J.; Xu, Y.; Lesker, T.R.; Schönlein, M.; Konczalla, L.; Giannou, A.D.; Pelczar, P.; Kylies, D.; Puelles, V.G.; Bielecka, A.A.; et al. Microbiota-derived 3-IAA influences chemotherapy efficacy in pancreatic cancer. Nature 2023, 615, 168–174. [Google Scholar] [CrossRef]
  63. Tempero, M.A.; Malafa, M.P.; Al-Hawary, M.; Behrman, S.W.; Benson, A.B.; Cardin, D.B.; Chiorean, E.G.; Chung, V.; Czito, B.; Del Chiaro, M.; et al. Pancreatic Adenocarcinoma, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2021, 19, 439–457. [Google Scholar] [CrossRef]
  64. Pfisterer, N.; Lingens, C.; Heuer, C.; Dang, L.; Neesse, A.; Ammer-Herrmenau, C. The Microbiome in PDAC—Vantage Point for Future Therapies? Cancers 2022, 14, 5974. [Google Scholar] [CrossRef]
  65. Gori, S.; Inno, A.; Belluomini, L.; Bocus, P.; Bisoffi, Z.; Russo, A.; Arcaro, G. Gut microbiota and cancer: How gut microbiota modulates activity, efficacy and toxicity of antitumoral therapy. Crit. Rev. Oncol. Hematol. 2019, 143, 139–147. [Google Scholar] [CrossRef]
  66. 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, 159–168. [Google Scholar] [CrossRef]
  67. 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. [Google Scholar] [CrossRef]
  68. Ciernikova, S.; Novisedlakova, M.; Cholujova, D.; Stevurkova, V.; Mego, M. The Emerging Role of Microbiota and Microbiome in Pancreatic Ductal Adenocarcinoma. Biomedicines 2020, 8, 565. [Google Scholar] [CrossRef]
  69. Cheng, K.-W.; Tseng, C.-H.; Tzeng, C.-C.; Leu, Y.-L.; Cheng, T.-C.; Wang, J.-Y.; Chang, J.-M.; Lu, Y.-C.; Cheng, C.-M.; Chen, I.-J.; et al. Pharmacological inhibition of bacterial β-glucuronidase prevents irinotecan-induced diarrhea without impairing its antitumor efficacy in vivo. Pharmacol. Res. 2019, 139, 41–49. [Google Scholar] [CrossRef]
  70. Yue, B.; Gao, R.; Wang, Z.; Dou, W. Microbiota-Host-Irinotecan Axis: A New Insight Toward Irinotecan Chemotherapy. Front. Cell. Infect. Microbiol. 2021, 11, 710945. [Google Scholar] [CrossRef]
  71. Mahdy, M.S.; Azmy, A.F.; Dishisha, T.; Mohamed, W.R.; Ahmed, K.A.; Hassan, A.; El Aidy, S.; El-Gendy, A.O. Irinotecan-gut microbiota interactions and the capability of probiotics to mitigate Irinotecan-associated toxicity. BMC Microbiol. 2023, 23, 1–22. [Google Scholar] [CrossRef]
  72. Panebianco, C.; Pisati, F.; Villani, A.; Andolfo, A.; Ulaszewska, M.; Bellini, E.; Ferro, C.; Lombardi, R.; Orsenigo, F.; Latiano, T.P.; et al. Counteracting gemcitabine+nab-paclitaxel induced dysbiosis in KRAS wild type and KRASG12D mutated pancreatic cancer in vivo model. Cell Death Discov. 2023, 9, 1–11. [Google Scholar] [CrossRef]
  73. Gong, J.; Chehrazi-Raffle, A.; Placencio-Hickok, V.; Guan, M.; Hendifar, A.; Salgia, R. The gut microbiome and response to immune checkpoint inhibitors: Preclinical and clinical strategies. Clin. Transl. Med. 2019, 8, 9. [Google Scholar] [CrossRef]
  74. Bartolini, I.; Nannini, G.; Risaliti, M.; Matarazzo, F.; Moraldi, L.; Ringressi, M.N.; Taddei, A.; Amedei, A. Impact of microbiota-immunity axis in pancreatic cancer management. World J. Gastroenterol. 2022, 28, 4527–4539. [Google Scholar] [CrossRef]
  75. Chandra, V.; McAllister, F. Therapeutic potential of microbial modulation in pancreatic cancer. Gut 2021, 70, 1419–1425. [Google Scholar] [CrossRef]
  76. 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, 33–37.e6. [Google Scholar] [CrossRef]
  77. Geller, L.T.; Straussman, R. Intratumoral bacteria may elicit chemoresistance by metabolizing anticancer agents. Mol. Cell. Oncol. 2017, 5, e1405139. [Google Scholar] [CrossRef]
  78. Guenther, M.; Surendran, S.A.; Hauer, N.; Fahrenschon, F.; Arslan, M.D.; Ormanns, S. Intratumoral bacterial abundance confers poor response to adjuvant gemcitabine in resected pancreatic cancer patients which is mitigated by postoperative antibiotics. Br. J. Cancer 2025, 133, 574–581. [Google Scholar] [CrossRef]
  79. Corty, R.W.; Langworthy, B.W.; Fine, J.P.; Buse, J.B.; Sanoff, H.K.; Lund, J.L. Antibacterial Use Is Associated with an Increased Risk of Hematologic and Gastrointestinal Adverse Events in Patients Treated with Gemcitabine for Stage IV Pancreatic Cancer. Oncol. 2020, 25, 579–584. [Google Scholar] [CrossRef]
  80. Mohindroo, C.; Hasanov, M.; Rogers, J.E.; Dong, W.; Prakash, L.R.; Baydogan, S.; Mizrahi, J.D.; Overman, M.J.; Varadhachary, G.R.; Wolff, R.A.; et al. Antibiotic use influences outcomes in advanced pancreatic adenocarcinoma patients. Cancer Med. 2021, 10, 5041–5050. [Google Scholar] [CrossRef]
  81. Fulop, D.J.; Zylberberg, H.M.; Wu, Y.L.; Aronson, A.; Labiner, A.J.; Wisnivesky, J.; Cohen, D.J.; Sigel, K.M.; Lucas, A.L. Association of Antibiotic Receipt With Survival Among Patients With Metastatic Pancreatic Ductal Adenocarcinoma Receiving Chemotherapy. JAMA Netw. Open 2023, 6, e234254. [Google Scholar] [CrossRef]
  82. Hajishengallis, G.; Lamont, R.J. Beyond the red complex and into more complexity: The polymicrobial synergy and dysbiosis (PSD) model of periodontal disease etiology. Mol. Oral Microbiol. 2012, 27, 409–419. [Google Scholar] [CrossRef]
  83. Chen, S.-M.; Hsu, L.-J.; Lee, H.-L.; Lin, C.-P.; Huang, S.-W.; Lai, C.J.-L.; Lin, C.-W.; Chen, W.-T.; Chen, Y.-J.; Lin, Y.-C.; et al. Lactobacillus Attenuate the Progression of Pancreatic Cancer Promoted by Porphyromonas Gingivalis in K-rasG12D Transgenic Mice. Cancers 2020, 12, 3522. [Google Scholar] [CrossRef]
  84. Guan, S.-W.; Lin, Q.; Yu, H.-B. Intratumour microbiome of pancreatic cancer. World J. Gastrointest. Oncol. 2023, 15, 713–730. [Google Scholar] [CrossRef]
  85. Taper, H.S.; Roberfroid, M.B. Possible adjuvant cancer therapy by two prebiotics--inulin or oligofructose. In Vivo. 2005, 19, 201–204. [Google Scholar]
  86. Chen, S.-M.; Chieng, W.-W.; Huang, S.-W.; Hsu, L.-J.; Jan, M.-S. The synergistic tumor growth-inhibitory effect of probiotic Lactobacillus on transgenic mouse model of pancreatic cancer treated with gemcitabine. Sci. Rep. 2020, 10, 1–12. [Google Scholar] [CrossRef] [PubMed]
  87. Liang, Y.; Du, M.; Li, X.; Gao, J.; Li, Q.; Li, H.; Li, J.; Gao, X.; Cong, H.; Huang, Y.; et al. Upregulation of Lactobacillus spp. in gut microbiota as a novel mechanism for environmental eustress-induced anti-pancreatic cancer effects. Gut Microbes 2025, 17, 2470372. [Google Scholar] [CrossRef] [PubMed]
  88. Mousavi, S.H.; Zahid, S.U.; Wardak, K.; Azimi, K.A.; Hosseini, S.M.R.; Wafaee, M.; Dhama, K.; Sah, R.; Rabaan, A.A.; Arteaga-Livias, K.; et al. Mapping the Changes on Incidence, Case Fatality Rates and Recovery Proportion of COVID-19 in Afghanistan Using Geographical Information Systems. Arch. Med Res. 2020, 51, 600–602. [Google Scholar] [CrossRef] [PubMed]
  89. Longhi, G.; van Sinderen, D.; Ventura, M.; Turroni, F. Microbiota and Cancer: The Emerging Beneficial Role of Bifidobacteria in Cancer Immunotherapy. Front. Microbiol. 2020, 11, 575072. [Google Scholar] [CrossRef]
  90. Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.-L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef]
  91. Maher, S.; Elmeligy, H.A.; Aboushousha, T.; Helal, N.S.; Ossama, Y.; Rady, M.; Hassan, A.M.A.; Kamel, M. Synergistic immunomodulatory effect of synbiotics pre- and postoperative resection of pancreatic ductal adenocarcinoma: A randomized controlled study. Cancer Immunol. Immunother. 2024, 73, 1–12. [Google Scholar] [CrossRef]
  92. Zhao, X.; Zhao, J.; Li, D.; Yang, H.; Chen, C.; Qin, M.; Wen, Z.; He, Z.; Xu, L. Akkermansia muciniphila: A potential target and pending issues for oncotherapy. Pharmacol. Res. 2023, 196, 106916. [Google Scholar] [CrossRef] [PubMed]
  93. Tabrizi, E.; Tabrizi, F.P.F.; Khaled, G.M.; Sestito, M.P.; Jamie, S.; Boone, B.A. Unraveling the gut microbiome’s contribution to pancreatic ductal adenocarcinoma: Mechanistic insights and therapeutic perspectives. Front. Immunol. 2024, 15, 1434771. [Google Scholar] [CrossRef] [PubMed]
  94. Trone, K.; Rahman, S.; Green, C.H.; Venegas, C.; Martindale, R.; Stroud, A. Synbiotics and Surgery: Can Prebiotics and Probiotics Affect Inflammatory Surgical Outcomes? Curr. Nutr. Rep. 2023, 12, 238–246. [Google Scholar] [CrossRef] [PubMed]
  95. Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023, 15, 2211. [Google Scholar] [CrossRef]
  96. Qiao, C.; Wang, L.; Huang, C.; Jia, Q.; Bao, W.; Guo, P.; Tan, D.; Chen, Z.; Shi, C.; Rao, Z.; et al. Engineered Bacteria Manipulate Cysteine Metabolism to Boost Ferroptosis-Based Pancreatic Ductal Adenocarcinoma Therapy. Adv. Mater. 2024, 37, e2412982. [Google Scholar] [CrossRef]
  97. Xu, X.; Ding, Y.; Dong, Y.; Yuan, H.; Xia, P.; Qu, C.; Ma, J.; Wang, H.; Zhang, X.; Zhao, L.; et al. Nanobody-Engineered Biohybrid Bacteria Targeting Gastrointestinal Cancers Induce Robust STING-Mediated Anti-Tumor Immunity. Adv. Sci. 2024, 11, e2401905. [Google Scholar] [CrossRef]
  98. Tan, W.; Duong, M.T.-Q.; Zuo, C.; Qin, Y.; Zhang, Y.; Guo, Y.; Hong, Y.; Zheng, J.H.; Min, J.-J. Targeting of pancreatic cancer cells and stromal cells using engineered oncolytic Salmonella typhimurium. Mol. Ther. 2022, 30, 662–671. [Google Scholar] [CrossRef]
  99. Han, Z.-Y.; Fu, Z.-J.; Wang, Y.-Z.; Zhang, C.; Chen, Q.-W.; An, J.-X.; Zhang, X.-Z. Probiotics functionalized with a gallium-polyphenol network modulate the intratumor microbiota and promote anti-tumor immune responses in pancreatic cancer. Nat. Commun. 2024, 15, 1–18. [Google Scholar] [CrossRef]
  100. Asar, M.C.; Franco, A.; Soendergaard, M. Phage Display Selection, Identification, and Characterization of Novel Pancreatic Cancer Targeting Peptides. Biomolecules 2020, 10, 714. [Google Scholar] [CrossRef]
  101. Bedi, D.; Gillespie, J.W.; Petrenko, V.A. Selection of pancreatic cancer cell-binding landscape phages and their use in development of anticancer nanomedicines. Protein Eng. Des. Sel. 2014, 27, 235–243. [Google Scholar] [CrossRef]
  102. Wang, T.; Narayanaswamy, R.; Ren, H.; Gillespie, J.W.; Petrenko, V.A.; Torchilin, V.P. Phage-derived protein-mediated targeted chemotherapy of pancreatic cancer. J. Drug Target. 2017, 26, 505–515. [Google Scholar] [CrossRef]
  103. Eissa, I.R.; Bustos-Villalobos, I.; Ichinose, T.; Matsumura, S.; Naoe, Y.; Miyajima, N.; Morimoto, D.; Mukoyama, N.; Zhiwen, W.; Tanaka, M.; et al. The Current Status and Future Prospects of Oncolytic Viruses in Clinical Trials against Melanoma, Glioma, Pancreatic, and Breast Cancers. Cancers 2018, 10, 356. [Google Scholar] [CrossRef] [PubMed]
  104. Rahal, A.; Musher, B. Oncolytic viral therapy for pancreatic cancer. J. Surg. Oncol. 2017, 116, 94–103. [Google Scholar] [CrossRef] [PubMed]
  105. Yokoda, R.T.; Nagalo, B.M.; Borad, M.J. Oncolytic Adenoviruses in Gastrointestinal Cancers. Biomedicines 2018, 6, 33. [Google Scholar] [CrossRef] [PubMed]
  106. Freytag, S.O.; Barton, K.N.; Brown, S.L.; Narra, V.; Zhang, Y.; Tyson, D.; Nall, C.; Lu, M.; Ajlouni, M.; Movsas, B.; et al. Replication-competent Adenovirus-mediated Suicide Gene Therapy with Radiation in a Preclinical Model of Pancreatic Cancer. Mol. Ther. 2007, 15, 1600–1606. [Google Scholar] [CrossRef]
  107. Yokoda, R.; Nagalo, B.M.; Arora, M.; Egan, J.B.; Bogenberger, J.M.; DeLeon, T.T.; Zhou, Y.; Ahn, D.H.; Borad, M.J. Oncolytic virotherapy in upper gastrointestinal tract cancers. Oncolytic Virotherapy 2018, 7, 13–24. [Google Scholar] [CrossRef]
  108. Gayral, M.; Lulka, H.; Hanoun, N.; Biollay, C.; Sèlves, J.; Vignolle-Vidoni, A.; Berthommé, H.; Trempat, P.; Epstein, A.L.; Buscail, L.; et al. Targeted Oncolytic Herpes Simplex Virus Type 1 Eradicates Experimental Pancreatic Tumors. Hum. Gene Ther. 2015, 26, 104–113. [Google Scholar] [CrossRef]
  109. Eissa, I.R.; Naoe, Y.; Bustos-Villalobos, I.; Ichinose, T.; Tanaka, M.; Zhiwen, W.; Mukoyama, N.; Morimoto, T.; Miyajima, N.; Hitoki, H.; et al. Genomic Signature of the Natural Oncolytic Herpes Simplex Virus HF10 and Its Therapeutic Role in Preclinical and Clinical Trials. Front. Oncol. 2017, 7, 149. [Google Scholar] [CrossRef]
  110. Yu, Y.A.; Galanis, C.; Woo, Y.; Chen, N.; Zhang, Q.; Fong, Y.; Szalay, A.A. Regression of human pancreatic tumor xenografts in mice after a single systemic injection of recombinant vaccinia virus GLV-1h68. Mol. Cancer Ther. 2009, 8, 141–151. [Google Scholar] [CrossRef]
  111. Dai, M.; Liu, S.; Chen, N.; Zhang, T.; You, L.; Zhang, F.Q.; Chou, T.; Szalay, A.; Fong, Y.; Zhao, Y. Oncolytic vaccinia virus in combination with radiation shows synergistic antitumor efficacy in pancreatic cancer. Cancer Lett. 2014, 344, 282–290. [Google Scholar] [CrossRef]
  112. Al Yaghchi, C.; Zhang, Z.; Alusi, G.; Lemoine, N.R.; Wang, Y. Vaccinia Virus, a Promising New Therapeutic Agent for Pancreatic Cancer. Immunotherapy 2015, 7, 1249–1258. [Google Scholar] [CrossRef] [PubMed]
  113. Mahalingam, D.; Wilkinson, G.A.; Eng, K.H.; Fields, P.; Raber, P.; Moseley, J.L.; Cheetham, K.; Coffey, M.; Nuovo, G.; Kalinski, P.; et al. Pembrolizumab in Combination with the Oncolytic Virus Pelareorep and Chemotherapy in Patients with Advanced Pancreatic Adenocarcinoma: A Phase Ib Study. Clin. Cancer Res. 2020, 26, 71–81. [Google Scholar] [CrossRef] [PubMed]
  114. Geisler, A.; Dieringer, B.; Elsner, L.; Klopfleisch, R.; Kurreck, J.; Fechner, H. Oncolytic Coxsackievirus B3 Strain PD-H Is Effective Against a Broad Spectrum of Pancreatic Cancer Cell Lines and Induces a Growth Delay in Pancreatic KPC Cell Tumors In Vivo. Int. J. Mol. Sci. 2024, 25, 11224. [Google Scholar] [CrossRef] [PubMed]
  115. Mahmoudian, F.; Gheshlagh, S.R.; Hemati, M.; Farhadi, S.; Eslami, M. The influence of microbiota on the efficacy and toxicity of immunotherapy in cancer treatment. Mol. Biol. Rep. 2024, 52, 1–14. [Google Scholar] [CrossRef]
  116. Baruch, E.N.; 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 2020, 371, 602–609. [Google Scholar] [CrossRef]
  117. Kim, Y.; Kim, G.; Kim, S.; Cho, B.; Kim, S.-Y.; Do, E.-J.; Bae, D.-J.; Kim, S.; Kweon, M.-N.; Song, J.S.; et al. Fecal microbiota transplantation improves anti-PD-1 inhibitor efficacy in unresectable or metastatic solid cancers refractory to anti-PD-1 inhibitor. Cell Host Microbe 2024, 32, 1380–1393.e9. [Google Scholar] [CrossRef]
  118. Routy, B.; Lenehan, J.G.; Miller, W.H.; Jamal, R.; Messaoudene, M.; Daisley, B.A.; Hes, C.; Al, K.F.; Martinez-Gili, L.; Punčochář, M.; et al. Fecal microbiota transplantation plus anti-PD-1 immunotherapy in advanced melanoma: A phase I trial. Nat. Med. 2023, 29, 2121–2132. [Google Scholar] [CrossRef]
  119. Davar, D.; Dzutsev, A.K.; McCulloch, J.A.; Rodrigues, R.R.; Chauvin, J.-M.; Morrison, R.M.; Deblasio, R.N.; Menna, C.; Ding, Q.; Pagliano, O.; et al. Fecal microbiota transplant overcomes resistance to anti–PD-1 therapy in melanoma patients. Science 2021, 371, 595–602. [Google Scholar] [CrossRef]
  120. DeFilipp, Z.; Bloom, P.P.; Torres Soto, M.; Mansour, M.K.; Sater, M.R.A.; Huntley, M.H.; Turbett, S.; Chung, R.T.; Chen, Y.-B.; Hohmann, E.L. Drug-Resistant E. coli Bacteremia Transmitted by Fecal Microbiota Transplant. N. Engl. J. Med. 2019, 381, 2043–2050. [Google Scholar] [CrossRef]
  121. Halsey, T.M.; Thomas, A.S.; Hayase, T.; Ma, W.; Abu-Sbeih, H.; Sun, B.; Parra, E.R.; Jiang, Z.-D.; DuPont, H.L.; Sanchez, C.; et al. Microbiome alteration via fecal microbiota transplantation is effective for refractory immune checkpoint inhibitor–induced colitis. Sci. Transl. Med. 2023, 15, eabq4006. [Google Scholar] [CrossRef]
  122. Elkrief, A.; Waters, N.R.; Smith, N.; Dai, A.; Slingerland, J.; Aleynick, N.; Febles, B.; Gogia, P.; Socci, N.D.; Lumish, M.; et al. Immune-Related Colitis Is Associated with Fecal Microbial Dysbiosis and Can Be Mitigated by Fecal Microbiota Transplantation. Cancer Immunol. Res. 2023, 12, 308–321. [Google Scholar] [CrossRef] [PubMed]
  123. Wang, Y.; Wiesnoski, D.H.; Helmink, B.A.; Gopalakrishnan, V.; Choi, K.; DuPont, H.L.; Jiang, Z.-D.; Abu-Sbeih, H.; Sanchez, C.A.; Chang, C.-C.; et al. Fecal microbiota transplantation for refractory immune checkpoint inhibitor-associated colitis. Nat. Med. 2018, 24, 1804–1808. [Google Scholar] [CrossRef]
  124. Chen, M.; Liu, M.; Li, C.; Peng, S.; Li, Y.; Xu, X.; Sun, M.; Sun, X. Fecal Microbiota Transplantation Effectively Cures a Patient With Severe Bleeding Immune Checkpoint Inhibitor-Associated Colitis and a Short Review. Front. Oncol. 2022, 12, 913217. [Google Scholar] [CrossRef]
  125. Ding, X.; Li, Q.; Li, P.; Chen, X.; Xiang, L.; Bi, L.; Zhu, J.; Huang, X.; Cui, B.; Zhang, F. Fecal microbiota transplantation: A promising treatment for radiation enteritis? Radiother. Oncol. 2020, 143, 12–18. [Google Scholar] [CrossRef]
  126. Cui, J.Q.; Tian, H.L.; Wang, X.J.; Wang, L.; Liu, Y.K.; Ye, C.; Ding, L.F.; Li, N.; Chen, Q.Y. [Analysis of short-term efficacy of perioperative fecal microbiota transplantation combined with nutritional support in patients with radiation-induced enteritis complicated by intestinal obstruction]. Chin. J. Gastrointest. Surg. 2023, 26, 955–962. [Google Scholar] [CrossRef]
Table 1. Microbiota and Therapy in PDAC.
Table 1. Microbiota and Therapy in PDAC.
Author/StudyMicroorganism/MetaboliteDrug/TherapyMechanismEffect on Therapy
Geller et al. 2017 [54]M. hyorhinis; GammaproteobacteriaGemcitabineGemcitabine inactivation via cytidine deaminase (CDD)Resistance
Weniger et al. 2021 [55]K. pneumoniaeGemcitabinePresence associated with reduced PFS under gemcitabineResistance
Lehouritis et al. 2015 [56]E. coli (non-pathogenic strain)Gemcitabine and othersDrug structural modification, leading to resistanceResistance
Ye et al., Udayasuryan et al., Yu et al. 2017, 2022, 2024 [57,58,59]F. nucleatum5-FUTLR4/MYD88-dependent inhibition of apoptosisResistance
Bai et al. 2023 [60]B. fragilis (non-toxigenic)OxaliplatinT-cell activation enhances oxaliplatin-induced apoptosisSensitization
Bai et al., 2023 Panebianco et al. 2022 [60,61]Butyrate-producing bacteriaOxaliplatinCD8+ T-cell activation; depletion increases resistanceSensitization
Panebianco et al. 2022 [61]ButyrateGemcitabineInduction of apoptosis in PDAC cellsSensitization
Tintelnot et al. 2023 [62]Indole-3-acetic acid (3-IAA)GemcitabineROS accumulation, impaired cancer metabolismSensitization
Pushalkar et al. 2018 [38]Multiple species (antibiotic-sensitive)Anti-PD-1 immunotherapyMicrobial ablation enhances PD-1 blockade efficacySensitization
CDD: Cytidine deaminase, PFS: Progression-free survival, TLR4: Toll-like receptor 4, MYD88: Myeloid differentiation primary response 88, PDAC: Pancreatic ductal adenocarcinoma, ROS: Reactive oxygen species, PD-1: Programmed cell death protein 1.
Table 2. Gut Microbiota and Chemotherapy-Related Toxicities in PDAC.
Table 2. Gut Microbiota and Chemotherapy-Related Toxicities in PDAC.
DrugMicroorganism/MetaboliteToxicityMechanismIntervention/Findings
5-Fluorouracil Escherichia, Enterococcus spp., ClostridiumMucositis, bacteremia, sepsisPathogenic overgrowth predisposes to infectionsAssociation with dysbiosis 2017, 2018 [66,67]
CisplatinLachnospiraceae, Lactobacillus (protective, promoted by D-methionine)General toxicity (attenuated by microbiota modulation)D-methionine fosters beneficial taxa, reducing toxicityD-methionine promotes protective taxa 2017, 2018 [66,67]
IrinotecanProteobacteria, Clostridium clusters, Fusobacteria (β-glucuronidase producers)Severe diarrhea, mucosal injuryβ-glucuronidase hydrolyzes SN-38G back to SN-38, damaging mucosaTargeted GUS inhibitors and probiotics reduce toxicity 2020, 2019, 2021, 2023 [68,69,70,71]
Gemcitabine + Nab-paclitaxelSCFA-producing bacteria (butyrate producers)Dysbiosis, chemotherapy side effects, stromatogenesisLoss of SCFA producers; restored with probiotics, improving diversityProbiotic supplementation mitigates side effects, restores SCFA producers 2023 [72]
GUS: β-glucuronidase, SN-38: Active irinotecan metabolite, SN-38G: SN-38 glucuronide, SCFA: Short-chain fatty acid.
Table 3. Antibiotics and Microbiota Modulation in PDAC Therapy.
Table 3. Antibiotics and Microbiota Modulation in PDAC Therapy.
Study/AuthorModel/SettingAntibiotic/InterventionMechanism/FindingOutcome
Sethi et al. 2018 [76]Subcutaneous PDAC (murine)Broad-spectrum antibiotics↑ IFN-γ cytotoxic T cells; ↓ IL-17A/IL-10 T cellsDelayed tumor growth (preclinical)
Pushalkar et al. 2018 [38]Orthotopic PDAC (murine)Broad-spectrum antibiotics↑ M1 macrophages, CD4+ Th1, CD8+; ↓ TLR2/5 signalingReduced tumor burden (preclinical)
Weniger et al. 2021[55]Preclinical/human tissueCiprofloxacin (counteracts Gammaproteobacteria)Reverses Gammaproteobacteria-mediated gemcitabine resistanceRestored gemcitabine efficacy
Fulop et al. 2023 [81]Clinical cohort, gemcitabine+nab-paclitaxelAntibiotics during GnP treatmentImproved PFS (5.8 vs 2.7 months); better outcomesClinical survival benefit
Weniger et al. 2021 [55]Clinical cohort, bile culture (K. pneumoniae)Absence of Klebsiella pneumoniaeBetter PFS when K. pneumoniae is absentImproved progression-free survival
Hajishengallis et al. 2012 [82]Clinical retrospective cohortAntibiotics + Gemcitabine vs FluoropyrimidinesGemcitabine benefits, not with fluoropyrimidinesImproved OS/PFS with gemcitabine
Fulop et al. 2023 [81]Clinical metastatic PDACMacrolides and quinolonesSurvival benefit of +2–3 monthsImproved OS/PFS in metastatic PDAC
Geller et al. 2017 [54]Murine colon carcinoma modelLocal release of ampicillin, chloramphenicolTumor inhibition, apoptosis ↑; synergy with gemcitabineReduced chemoresistance; local effect with less systemic toxicity
PDAC: Pancreatic ductal adenocarcinoma, IFN: Interferon, IL: Interleukin, CD: Cluster of differentiation, Th: T helper, TLR2/5: Toll-like receptor 2/5, GnP: Gemcitabine/nab-paclitaxel, PFS: Progression-free survival, OS: Overall survival.
Table 4. Probiotics, Synbiotics, and Postbiotics in PDAC.
Table 4. Probiotics, Synbiotics, and Postbiotics in PDAC.
Species/MoleculeMechanismTherapeutic EffectModel/Setting
Lactobacillus paracasei, L. reuteri, L. rhamnosusp53 activation, cell cycle arrest, EMT modulationAntitumor activity; enhances gemcitabine efficacyPreclinical models 2005, 2020 [83,85,86]
Lactobacillus (co-administered with gemcitabine)Reduced PanIN progression; improved hepatic enzyme profileImproved drug tolerability and efficacyMouse models 2020 [86]
Lactobacillus reuteriPromotes NK cell infiltration; correlated with improved survivalSuppression of PDAC progression; survival benefitMouse and clinical correlations 2025 [87]
Ferrichrome (from L. casei)p53-mediated apoptosis, including in 5-FU-resistant cellsAntineoplastic effectIn vitro models 2020 [88]
Bifidobacterium longumEnhances CD8+ T-cell infiltration; reduces immunosuppressive subsets; synergizes with anti-PD-L1Attenuates PDAC progression; potentiates immunotherapyPreclinical models 2020, 2015, 2024 [89,90,91]
Akkermansia muciniphilaBoosts CD8+ T-cell immunity; enhances checkpoint inhibitor efficacyImproved immunotherapy responsePreclinical and translational 2023, 2024 [92,93]
Synbiotics (probiotics + inulin)↑ CD8+ T-cell infiltration and IFN-γ; ↓ IL-1β, IL-6, IL-10Reduced postoperative complications and bacteremiaRandomized clinical trial (90 patients) 2024, 2023 [91,94]
Butyrate (SCFA)Enhances gemcitabine efficacy (oxidative stress, autophagy); HDAC inhibition; epigenetic regulationSuppresses tumor proliferation; potentiates chemotherapyPreclinical and mechanistic studies 2023 [62,95]
Indole-3-acetic acid Regulates tumor and immune cell activity; enhances chemotherapy responseEnhanced chemotherapy effectivenessPreclinical models 2023 [62]
EMT: Epithelial–mesenchymal transition, PanIN: Pancreatic intraepithelial neoplasia, NK: Natural killer, FU: Fluorouracil, CD: Cluster of differentiation, PD-L1: Programmed death-ligand 1, IFN: Interferon, IL: Interleukin, SCFA: Short-chain fatty acid, HDAC: Histone deacetylase.
Table 5. Engineered Bacteria as Therapeutic Tools in PDAC.
Table 5. Engineered Bacteria as Therapeutic Tools in PDAC.
Engineered BacteriumStrategyMechanismTherapeutic Effect
Hypoxia-responsive E. coliGenetically modified to secrete cyst(e)inase (CGL)Depletes cysteine/cystine pools → induces ferroptosis via lipid peroxidation; activates antitumor immunityFerroptosis induction and immune activation 2025 [96]
Synthetic Salmonella strains (OMVs with STING agonists)Delivery of STING agonists via outer membrane vesiclesEnhances immune infiltration; synergizes with checkpoint inhibitorsImproved immunotherapy efficacy 2024 [97]
Attenuated Salmonella Typhimurium (immunomodulatory proteins)Engineered to secrete immunomodulatory proteinsInhibits PDAC growth; alters cytokine profilesSuppression of PDAC growth 2022 [98]
Lactobacillus rhamnosus GG functionalized with Ga–polyphenol network (LGG@Ga-poly)Functionalization with gallium–polyphenol networkDepletes Proteobacteria and LPS; remodels tumor immune microenvironmentEnhanced immunotherapeutic response 2024 [99]
CGL: Cystathionine γ-lyase, OMVs: Outer membrane vesicles, STING: Stimulator of interferon genes, PDAC: Pancreatic ductal adenocarcinoma, Ga–polyphenol: Gallium–polyphenol complex, LPS: Lipopolysaccharide.
Table 6. Bacteriophages and Oncolytic Viruses in PDAC.
Table 6. Bacteriophages and Oncolytic Viruses in PDAC.
AgentStrategyMechanismTherapeutic Effect
Lytic bacteriophagesEradication of tumor-associated pathogenic bacteriaSelective bacterial lysisPotential elimination of pathogenic microbiota 2020 [100]
Phage peptide MCA1Phage display-identified peptide binds PDAC cellsHigh-specificity binding with nanomolar affinityImaging and targeted drug delivery 2020 [100]
Phage-decorated liposomes (doxorubicin)Fusion proteins used to decorate liposomesEnhanced tumor selectivity and cytotoxicityEnhanced doxorubicin efficacy 2014 [101]
Phage-functionalized micelles (paclitaxel)Micelles functionalized with phage proteinsImproved drug targeting and solubility; reduced off-target toxicityImproved paclitaxel delivery and safety, 2018 [102]
Adenoviruses (first-generation ONYX-015)First oncolytic adenovirus vectorLimited replication; immune clearanceLimited efficacy 2018. 2017 [103,104]
Next-generation adenovirusesEngineered to overcome antiviral resistance, radiosensitize, and remodel stromaEnhanced persistence, radiosensitivity, and stromal modulationImproved antitumor activity 2018. 2007 [105,106,107]
HSV-based oncolytic virusesEngineered or naturally mutated strainsInduces apoptosis/necrosis; stimulates immune activationSuppressed PDAC growth; synergy with gemcitabine 2015, 2017 [108,109]
Vaccinia virus strainsEngineered or naturally attenuated strainsDirect oncolysis; synergizes with chemo/radiotherapyPreclinical efficacy; ongoing clinical trials 2009. 2014, 2015 [110,111,112]
Reoviruses (Pelareorep)Exploits replication in RAS-mutant cellsReplicates in RAS-mutant cells; immunotherapy synergyLimited monotherapy efficacy; synergy with ICIs 2020 [113]
Coxsackievirus B3 (PD-H strain)Naturally lytic strain targeting PDACPotent lytic activity; requires stromal-targeting combinationSuppressed tumor growth in vivo 2024 [114]
MCA1: Mia PaCa-2 Cell-binding peptide 1, PDAC: Pancreatic ductal adenocarcinoma, HSV: Herpes simplex virus, RAS: Rat sarcoma virus, ICIs: Immune checkpoint inhibitors.
Table 7. Fecal Microbiota Transplantation in PDAC.
Table 7. Fecal Microbiota Transplantation in PDAC.
Study/SettingDonor/SourceOutcome
Chandra et al. 2021 [75]Immunotherapy responders or long-term PDAC survivorsReduced tumor burden; ↑ T-cell infiltration
Riquelme et al. 2019 [40]Long-term PDAC survivors (diverse intratumoral microbiota)Higher intratumoral diversity correlated with survival (Pseudoxanthomonas, Streptomyces, Saccharopolyspora, B. clausii)
Tintelnot et al. 2023 [62]Chemotherapy responders or healthy donorsImproved chemosensitivity; reduced tumor progression
No Melanoma trials (anti-PD-1 refractory) 2021, 2023, 2024 [116,117,118,119]Responder or healthy donorsReversal of anti-PD-1 resistance; partial and complete responses
De Filipp et al. 2019 [120]Shared donor (safety incident)Fatal bacteremia due to ESBL-producing E. coli
ICI-induced colitis (steroid/biologic-refractory) 2023, 2024, 2018, 2022 [121,122,123,124]Responder-derived or healthy donor FMTImproved colitis control; enabled ICI continuation
Ding et al. 2020 [125]FMT donors (clinical setting)Clinical improvement without serious adverse events
Cui et al. 2023 [126]FMT donors (extended follow-up)Improved GI quality of life and nutritional status (6 months)
PDAC: Pancreatic ductal adenocarcinoma, PD-1: Programmed cell death protein 1, ESBL: Extended-spectrum beta-lactamases, ICI: Immune checkpoint inhibitor, FMT: Fecal microbiota transplantation, GI: Gastrointestinal.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

De Lucia, S.S.; Nista, E.C.; Candelli, M.; Archilei, S.; Deutschbein, F.; Capuano, E.; Gasbarrini, A.; Franceschi, F.; Pignataro, G. Microbiota and Pancreatic Cancer: New Therapeutic Frontiers Between Engineered Microbes, Metabolites and Non-Bacterial Components. Cancers 2025, 17, 3618. https://doi.org/10.3390/cancers17223618

AMA Style

De Lucia SS, Nista EC, Candelli M, Archilei S, Deutschbein F, Capuano E, Gasbarrini A, Franceschi F, Pignataro G. Microbiota and Pancreatic Cancer: New Therapeutic Frontiers Between Engineered Microbes, Metabolites and Non-Bacterial Components. Cancers. 2025; 17(22):3618. https://doi.org/10.3390/cancers17223618

Chicago/Turabian Style

De Lucia, Sara Sofia, Enrico Celestino Nista, Marcello Candelli, Sebastiano Archilei, Franziska Deutschbein, Enrico Capuano, Antonio Gasbarrini, Francesco Franceschi, and Giulia Pignataro. 2025. "Microbiota and Pancreatic Cancer: New Therapeutic Frontiers Between Engineered Microbes, Metabolites and Non-Bacterial Components" Cancers 17, no. 22: 3618. https://doi.org/10.3390/cancers17223618

APA Style

De Lucia, S. S., Nista, E. C., Candelli, M., Archilei, S., Deutschbein, F., Capuano, E., Gasbarrini, A., Franceschi, F., & Pignataro, G. (2025). Microbiota and Pancreatic Cancer: New Therapeutic Frontiers Between Engineered Microbes, Metabolites and Non-Bacterial Components. Cancers, 17(22), 3618. https://doi.org/10.3390/cancers17223618

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

Article Metrics

Back to TopTop