Next Article in Journal
SERPINA3: Stimulator or Inhibitor of Pathological Changes
Next Article in Special Issue
In Vitro Evaluation of the Most Active Probiotic Strains Able to Improve the Intestinal Barrier Functions and to Prevent Inflammatory Diseases of the Gastrointestinal System
Previous Article in Journal
TGF-β1 and TGFβR2 Gene Polymorphisms in Patients with Unstable Angina
Previous Article in Special Issue
Fecal Microbiota Transplantation Could Improve Chronic Diarrhea in Cynomolgus Monkey by Alleviating Inflammation and Modulating Gut Microbiota
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pancreatic Cancer Resistance to Treatment: The Role of Microbiota

1
Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
2
Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2023, 11(1), 157; https://doi.org/10.3390/biomedicines11010157
Submission received: 29 November 2022 / Revised: 30 December 2022 / Accepted: 4 January 2023 / Published: 7 January 2023
(This article belongs to the Special Issue Advanced Research of Gut Microbiota in Health and Diseases)

Abstract

:
Pancreatic cancer (PC) is an aggressive malignancy and the fourth leading cause of cancer death in the United States and Europe. It is estimated that PC will be the second leading cause of cancer death by 2030. In addition to late diagnosis, treatment resistance is a major cause of shortened survival in pancreatic cancer. In this context, there is growing evidence that microbes play a regulatory role, particularly in therapy resistance and in creating a microenvironment in the tumor, that favors cancer progression. The presence of certain bacteria belonging to the gamma-proteobacteria or mycoplasmas appears to be associated with both pharmacokinetic and pharmacodynamic changes. Recent evidence suggests that the microbiota may also play a role in resistance mechanisms to immunotherapy and radiotherapy. However, the interactions between microbiota and therapy are bilateral and modulate therapy tolerance. Future perspectives are increasingly focused on elucidating the role of the microbiota in tumorigenesis and processes of therapy resistance, and a better understanding of these mechanisms may provide important opportunities to improve survival in these patients.

1. Pancreatic Cancer

Pancreatic cancer (PC) is an aggressive malignant tumor and the fourth leading cause of cancer death in the U.S. and Europe. PC kills nearly 50,000 people each year in North America alone. In fact, PC has the lowest survival rates for all stages combined (11%) among malignancies, and although the incidence is relatively stable, it is estimated to be the second leading cause of death by 2030 [1]. Because the symptoms of PC are usually nonspecific, the disease is usually not diagnosed until the late stages, when the patient complains of abdominal pain, unexplained weight loss, fatigue, nausea, or jaundice. Among exocrine tumors of the pancreas, which account for 95% of PC, pancreatic ductal adenocarcinoma (PDAC) is the most common and accounts for almost all pancreatic malignancies [2]. PDAC usually arises from the neoplastic transformation of pancreatic intraepithelial neoplasms (PanINs) and intraductal papillary mucinous neoplasms (IPMNs), typically found in 2% of the population, and is more common in people older than 70 years [2]. Several risk factors are significantly associated with the development of PC. For example, advanced age (71 years), male gender, heavy alcohol consumption, obesity, low vegetable diet, chronic pancreatitis (CP), diabetes, smoking, and family history have been shown to increase the risk [3]. In particular, cigarette smoking appears to be the cause of nearly 20% of pancreatic tumors [1], and current and former smokers have a 1.56- and 1.15-fold increased risk, respectively, according to a recent meta-analysis by Ben QW [4]. Having two or more first-degree relatives with PC is also a relevant risk factor, and it is estimated that approximately 8% of cases of PC are diagnosed in patients with a positive family history [5,6]. According to various meta-analyses, the prevalence of diabetes has increased threefold in PC patients. Moreover, this association is even higher in patients recently diagnosed with diabetes, which could be an early symptom of cancer [7].
Another risk factor is chronic pancreatitis, and analysis by Gandhi et al. estimated the standard incidence ratio (SIR) to be 22.61 in patients with chronic pancreatitis and as high as 63 in hereditary types [8]. By contrast, numerous other factors, such as coffee consumption [9], oral contraceptive use [10], and proton pump inhibitor use (PPI) [11], which were considered relevant additional risks in the past, showed no statistical association in further analyses. Tumorigenesis results directly and indirectly from alterations in various molecular pathways and is correlated with different genetic alterations, such as mutations in TP53, KRAS, CDKN2A, and SMAD4 [6]. However, genetic testing is currently indicated only for certain populations. BRCA2 genetic testing is recommended in patients with a family or personal history of breast cancer, with a family history of PC, and in individuals of Jewish descent. The presence of the CDKN2A mutation should be tested in patients with a family history of hereditary melanoma [12]. In addition to clinical examinations, the diagnostic process provides a complete blood analysis to monitor the indices of cholestasis and the presence of acute pancreatitis. The carbohydrate antigen 19-9 (CA 19-9) remains the most used serum tumor marker for PDAC, with a sensitivity of 80% among symptomatic patients. CT scans of the abdomen have a crucial role in diagnosing and staging the tumoral tissue. In addition to the CT technique, MRI or PET could also be included during the staging process, mostly in case of indeterminate and occult lesions [3]. Eventually, because of the high sensitivity and minimally invasive technique, endoscopic ultrasound (EUS) represents the first-choice modality for obtaining biopsies and definitive diagnosis [3].

1.1. Therapies for Pancreatic Cancer

In the PDAC treatment spectrum analysis, surgery is considered the only curative treatment for this neoplasm, but only a small percentage (10–20%) of patients can benefit from this treatment, as more than half of patients have the metastatic disease [1]. Adequate staging is required to differentiate patients with potentially resectable, borderline resectable, locally advanced, and metastatic disease [13]. In the early stages of cancer, the initiation of a neoadjuvant therapy protocol before surgery is still controversial [14]. The American Society of Clinical Oncology (ASCO) guidelines recommend that neoadjuvant therapy be reserved for patients with suspected extra pancreatic disease or radiographic evidence of mesenteric vessel infiltration who are at high surgical risk and have high serum levels of CA 19-9 without cholestasis [15]. The most commonly used strategy for resectable disease is surgical resection followed by adjuvant therapy. In this context, recent National Comprehensive Cancer Network (NCCN) guidelines recommend gemcitabine alone, gemcitabine in combination with capecitabine, continuous fluorouracil infusion (5 FU), or 5 FU/leucovorin therapy (NCCN) [16]. In borderline resectable and locally advanced diseases, no further surgery is recommended by the NCCN. At this stage, the choice is between chemotherapy or chemoradiotherapy as neoadjuvant therapy. For this purpose, FOLFIRINOX (a combination of folinic acid, fluorouracil, irinotecan, and oxaliplatinum), gemcitabine-nab-paclitaxel, and gemcitabine-cisplatin are currently approved as chemotherapy [15,16]. As previously reported, most patients are diagnosed with advanced-stage metastatic disease, with a median one-year survival rate of 7% [17]. Therapeutic strategies for metastatic disease include Nab-paclitaxel plus gemcitabine, FOLFIRINOX, gemcitabine and capecitabine, gemcitabine and erlotinib, gemcitabine, and cisplatin, or gemcitabine alone [14]. In addition to these therapies, the current interest in elucidating the molecular mechanisms and biology of cancer has led to the development of new targets and new modalities for PDAC treatment [14], which are summarized in Table 1. A panoramic of the novel treatments in development for PC is presented.

1.2. Microbiota and Pancreatic Cancer

There is growing evidence on how the gut microbiome, the genome of the entire community of microorganisms living in the gastrointestinal tract, can influence human health and disease [6]. New detection methods such as whole genome sequencing have facilitated the description and detection of an increasing number of bacteria, fungi, and viruses that can influence host inflammatory status, intestinal permeability, and carcinogenesis [18,19]. Gastrointestinal tumors, such as pancreatic ductal adenocarcinoma (PDAC), have a higher number of detections for type-specific microbiome fingerprints, mainly due to their spatial proximity [6,20]. The reciprocal balance between the pancreatic and gastrointestinal microbiota can lead to various pancreatic pathologies [21]. While various metabolites of the microbiota, such as short-chain fatty acids (SCFA), protect against tissue inflammation, and normal pancreatic β-cells secrete cathelicidin-related antimicrobial peptides (CRAMP) control pancreatic bacterial overgrowth [22]. In recent decades, several studies have highlighted the association between intestinal and oral dysbiosis and the presence of PC [23,24,25,26,27,28,29,30,31] (Table 2).
Dental disease and associated changes in the oral microbiome increase the risk for PDAC, as evidenced by decreased oral concentrations of Neisseria elongata and Streptococcus mitis and the increased abundance of Leptotrichia and Porphyromonas gingivalis in patients with PC [24, 25]. In recent decades, the presence of certain species, such as P. gingivalis or Helicobacter pylori, has been associated with higher rates of PC. Several pathological pathways have been proposed to explain this correlation. In particular, P. gingivalis may affect miRNA expression, immunological response, and arginine catabolism [26,27]. Furthermore, studies in animal models and in vitro have shown that P. gingivalis, Treponema denticola, and Tannerella forsythia can induce KRAS and p53 mutations and increase the risk of PC [28]. In addition to the luminal microbiota, other studies have demonstrated the physiological presence of a microbiome associated with pancreatic tissue. Specifically, bacterial cytidine deaminase (CDA) appears to be expressed four times more in PCs than in healthy controls [29], with a higher average abundance of the microbiome [18]. In general, the bacterial communities in the pancreas had a similar composition in terms of taxa as the oral microbiome. In addition, several studies have identified host variations in the genes involved in the immune response associated with different β-diversity [30]. Compared to healthy controls, PC patients showed an increased relative abundance of Porphyromonas, Prevotella, Bifidobacterium, and Synergistetes, as well as the phylum Euryarchaeota [18,31]. In another study, PDAC with KRAS mutations was associated with the enrichment of the genera Acinetobacter, Sphingopyxis, and Pseudomonas. PDAC tissues and fluid from IPMNs were enriched in Fusobacterium nucleatum, an oral pathogen [31], and depleted in Lactobacillus and phyla of Firmicutes and Proteobacteria [32], suggesting a role for Fusobacterium in the development of pancreatic neoplasia [33]. Bacterial communities may influence tumor progression through various metabolites such as SCFAs, gallic acid, lipopolysaccharide (LPS), and polyamines [34]. LPS is a common component of Gram-negative bacteria and could provide another pro-oncogenic stimulus by promoting local inflammation via the Toll-like receptor (TLR) [34,35] and subsequently mutating and activating the KRAS gene [34]. In addition, P53 mutations appear to be associated with a specific bacterial metabolite. In particular, gallic acid, produced by Bacillus subtilis and Lactobacillus Plantarum, was recently linked to pro-oncogenic Tpr53 mutations in a mouse model [36]. Numerous microorganisms can synthesize polyamines that are directly involved in cell growth [37]. These metabolites were significantly elevated in the mouse models of PC, and their reduction lead to a lower rate of cell anabolism [6,37]. In addition to bacterial communities, fungi have also been shown to be involved in the pathogenesis of PC [38]. Strikingly, the mycobiome of PDAC tissues is usually more pronounced, especially in the genera Malassezia and Alternaria, thus differing from the normal intestinal mycobiome. The pathogenic role of fungal components was mediated by the stimulation and secretion of IL-33 and activation of TH2 cells and ILC2. In mouse models, IL -33 was able to induce a chromatin switch that interacted with a genetic mutation such as KRAS [38]. In addition, glycans of fungal walls can also pursue oncogenic processes themselves by binding mannose-binding lectin (MBL) and recruiting the complement system [39]. Although antifungal treatments have appeared to halt carcinogenesis in previous in vitro experiments [40], they have not shown efficacy in animal models, likely due to the low concentrations of live fungi in tissues [41]. Several studies have found specific microbiome differences between patients with long-term survival (LTS) and short-term survival. In a study by Riquelme E et al., it was demonstrated that patients with a better prognosis had increased relative abundance of Alkalihalobacillus clausii (the new name of Bacillus clausii), Pseudoxanthomonas, Saccharopolyspora, and Streptomyces, with higher alpha diversity in their tissue-associated microbiome. Researchers transferring fecal microbiota transplantation (FMT) from LTS to mice also demonstrated the causal effect of these species in modulating tumor growth [40]. Moreover, the poorer prognosis was associated with the detection of intra-tumoral fusobacteria, although no specific differences in genetic mutations were found [23]. A subsequent analysis based on 187 PDAC patients also showed that certain strains were more prevalent in patients with metastases. Specifically, they found a higher abundance of Beutenbergia cavernae DSM 12333, Mycoplasma hypopneumoniae, Tolypothrix sp. PCC 7601, Acidovorax ebreuus TPSY, Agrobacterium radiobacter K84, Aggregatibacter aphorophilus NJ8700, Shigella sonnei Ss046, Salmonella enterica, Citrobacter freundii and the lower presence of Saccharomonospora Viridis DSM 43017 [42]. Although further studies are needed to clarify the clinical role of gut microbiota in pancreatic cancer, a recent promising study has demonstrated the importance of fecal microbiota analysis in diagnostic evaluations [43]. The authors considered 27 different species, such as Fusobacterium nucleatum/hwasookii, Alloscardovia omnicolens, Veillonella atypica, Faecalibacterium prausnitzii, Bifidobacterium bifidum Romboutsia timonensis, Bacteroides coprocola, Veillonella atypica, Methanobrevibacter smithii, Bacteroides finegoldii, and Alloscardovia omnicolens, noted an ability to discriminate between the disease and health of 84%, and of 94% when combined with serum CA 19-9 [43].

1.3. Microbiota and Chemotherapy

Therapy resistance is one of the main causes of shortened survival in pancreatic cancer [41]. Among the various factors involved in resistance, the gut microbiota seems to play an important role, mainly affecting drug metabolism and absorption [44,45] (Figure 1).
Interestingly, some bacteria belonging to the gamma-proteobacteria class, such as Escherichia coli [46], can interfere with gemcitabine metabolism by converting this drug to its inactive form (2,2-difluorodeoxycytidine to 2,2-difluorodeoxyuridine) via their long isoform of the bacterial enzyme CDA [29].
In addition, gamma-proteobacteria were detected at elevated levels in human PDAC. (29) Furthermore, other genera, such as Mycoplasma, can produce cytidine deaminase, and the use of tetrahydrouridine (THU), an inhibitor of CDA, can restore the normal anticancer effects of gemcitabine [47]. These effects have been extensively studied in colorectal cancer, and there are emerging data on PC. For example, colon cancer cells were found to develop chemotherapy resistance when cultured with bacteria derived from human pancreatic cancer, which are normally enriched in proteobacteria [48]. Moreover, in a recent study, the presence of K. pneumonia in the bile ducts was associated with a worse prognosis in the population treated with adjuvant therapy and gemcitabine, and a higher survival rate was observed after antibiotic therapy with quinolones [49]. On the other hand, the increased microbial production of vitamin K2 (menaquinone), usually detected in diabetes models, has also been associated with resistance to gemcitabine/paclitaxel [50]. As a result, the hypothesis of anti-oxidative cell protection against the accumulation of reactive oxygen species induced by chemotherapy has been put forward [50]. Another drug class of interest for microbiota-mediated chemoresistance is fluoropyrimidines, which are included in the FOLFIRINOX protocol [51]. As mentioned above, some studies have shown an increased inactivation rate of 5 FU sustained by Mycoplasma hyorhinis [52] and by Fusobacterium nucleatum (F. nucleatum) through the alteration of TLR4/MYD88-dependent autophagic metabolism, and the inhibition of 5-FU-induced cell apoptosis. Furthermore, the abundance of F. nucleatum has been linked to poor prognosis in pancreatic cancer [40,53,54]. Although no specific enzyme or mechanism has yet been identified [29], F. nucleatum has also been associated with resistance to oxaliplatin [54]. By contrast, some species, such as Bacteroides ovatus and Bacteroides xylanisolven, have been associated with enhanced recruitment of T cells and, consequently, with the enhancement of the effect of erlotinib: an EGFR tyrosine kinase inhibitor [55].

1.4. Microbiota and Immunotherapy

Immunotherapy is considered an effective therapeutic strategy for many neoplasms, such as melanoma and non-small cell lung carcinoma [56,57]. Immunotherapies mainly use monoclonal antibodies to stimulate an anti-tumor immune response directed against programmed cell death protein 1 (PD-1)/programmed death ligand 1 (PD-L1) or cytotoxic T lymphocyte antigen 4 (CTLA-4). Unfortunately, these strategies have not yet proven effective in PDAC [58], likely due to the specific characteristics of the tumor microenvironment (TME). The complex organization of the extracellular matrix, fibroblasts, and immunosuppressive leukocytes creates a difficult-to-target environment that promotes tumor growth [59]. An important role in this complex mechanism of resistance is also attributed to the microbiota. Recent evidence has shown that microbes influence the response to immunotherapy, regulate immune checkpoints, and promote cancer cell escape from the immune system [60,61,62]. The use of antibiotics in PDAC mouse models confirmed these findings and led to profound changes in the immune system, such as an increase in effector T cells and a change in tumor-associated macrophages (TAM) [18,62]. Other relevant changes included a higher proportion of antitumor cell types, such as Th1 CD8, and the release of gamma interferon and TNF alfa, as well as a decreased distribution of Treg cells and the production of IL-17 [63]. Notably, bacterial suppression has been associated with the increased expression of PD-1 and increased efficacy of checkpoint-targeted immunotherapy, with a synergistic effect on tumor size and an increase in T-cell activity [18]. Countervailing evidence is that the effect of antibiotics on reducing tumor growth did not occur in Rag1 knockout mice lacking mature T (and B) lymphocytes. This suggests that the immune system and microbiota must be involved in this process and that it is not a direct cytotoxic effect of the drug [61]. Moreover, the microbiota can contrast the effect of antibiotics by blocking IL 17, which, together with CD4 and th17 cells, is an important pro-carcinogenic factor in TME. [61]. A relatively new approach to modulating the gut microbiota is FMT, and a better prognosis and response to immunotherapy was observed in PC animal models that received stools from healthy donors or long-term survivors (LTS). Conversely, the use of antibiotics after FMT from healthy controls did not affect survival [39]. Other elements that appear to play a role in the immune evasion of PDAC are microRNAs, which have also been associated with increased tumor growth and resistance to therapy. MicroRNAs have been shown to alter pancreatic cell gene expression and influence immune responses, and recent studies have linked some bacteria to the expression of such miRNAs, suggesting additional pathways of immune regulation [64]. In a recent study, it was confirmed that the presence of megasphaera, capable of producing SCFA in tissues, showed a better response to anti-PD-1 therapies [65]. At present, promising preclinical model results have been achieved from the combination of anti-PD1 and antibiotics. On the contrary, clinical trials to evaluate the combination of pembrolizumab (NCT03637803) with lyophilized bacteria, or the combination of probiotics, vancomycin, and nivolumab (NCT03785210), are still in progress [18].

1.5. Microbiota and Radiotherapy

Radiotherapy (RT) is a therapeutic strategy based on ionizing radiation that results in direct damage to DNA, the formation of reactive oxygen species (ROS), and reactive nitrogen species (RNS) [66]. Irradiation can induce an intense immune response leading to tumor cell death and the release of antigens capable of stimulating cells of the immune system, such as CD8 T lymphocytes, resulting in a significant antitumor effect capable of killing tumor cells in distant, non-irradiated parts of the organism. This effect is known as the abscopal effect [60]. RT has been shown to be useful in resected PDAC patients to improve local control as an adjuvant therapy due to its cytostatic activity [51]. However, although the techniques are based on irradiating the tumor areas as selectively as possible, there are also effects on the surrounding healthy tissue, especially considering the anatomical location of the pancreas. In this context, a complex relationship between the microbiota and the effect or tolerance of radiotherapy has been recently identified [67]. In some studies with germ-free mice, increased resistance to the radiation effect has been described, both in terms of the therapeutic effect and toxicity [68]. Conversely, an enhanced effect of RT was observed after the administration of vancomycin in cancer models. This synergistic result was attributed to the elimination of Gram-positive bacteria, which can interfere with the presentation of tumor antigens to cytolytic CD8+ T cells [69]. Further evidence for the role of the microbiota as a predictor of the response to RT is provided by hepatocellular carcinoma (HCC). A recent study found that c-di-adenosine monophosphate (c-di-AMP), a bacterial metabolite, enhances the RT-related immune response [70]. Although these studies were not performed in pancreatic cancer, they justify the hypothesis of the influence of the microbiota on the immune system during RT treatments and thus provide the basis for future correlation and causality studies in this neoplasm.

1.6. Bidirectional Relationship between Therapy and Microbiota

In recent years, a bidirectional relationship between microbiota and therapeutic strategies in PDAC has been established. Both chemotherapy and radiotherapy can lead to alterations in the gut microbiota, which may limit its tolerability and influence its toxicity [63]. As mentioned earlier, radiation can cause tissue damage leading to enteritis [71], and the gut microbiota appears to play a central role in these processes. The most commonly observed changes after irradiation are the decrease in Lactobacillus spp. and Bifidobacterium spp. and the increase in Escherichia coli and Staphylococcus spp. [72]. These changes lead to the impairment of intestinal barrier integrity and the production of proinflammatory cytokines. In this regard, pretreatment with FMT or an antibiotic cocktail to restore the intestinal microenvironment after irradiation provides interesting results [73]. As for traditional chemotherapy, several studies are based on gemcitabine therapy. During treatment, an increased abundance of proinflammatory bacteria, such as Proteobacteria, and a decrease in butyrate-producing bacteria, such as Lachnospiraceae and Ruminococcaceae, have been described. These changes are associated with an increase in intestinal permeability, inflammation, and a decrease in the anticancer activity of butyrate, which promotes tumor progression [74,75,76]. In addition, a decrease in SCAF production has been noted during paclitaxel therapy, which has been associated with a higher rate of Clostridioides difficile infection [77] and an increase in Mucispirillium in the colon, which may play a role in neuroinflammation and cause peripheral neuropathy [78]. With respect to the drugs included in the FOLFIRINOX protocol, toxicities related to changes in the microbiota have been described for 5-FU as well as for oxaliplatin and irinotecan. Therapy with 5-FU is frequently compromised by the occurrence of mucositis, likely due to the depletion of mucin-producing bacteria such as Lactobacillus and Streptococcus [79]. Therefore, it has been reported that the administration of Lactobacillus and Bifidobacterium could increase the production of anti-inflammatory cytokines and thus reduce the intensity of the damage [80]. The efficacy of oxaliplatin is often limited by gastrointestinal toxicity and pain; interestingly, these effects are reduced in germ-free mice after antibiotic administration or after FMT [81]. The mechanisms involved are drug-induced increased levels of bacterial products, such as LPS, in the spinal ganglion, leading to inflammation [71]. In addition, the role of the microbiota in irinotecan-mediated diarrhea has recently been investigated [45]. In particular, this therapy seems to increase the abundance of Enterobacteriaceae, leading to an increase in bacterial β-glucuronidase (BGUS) and LPS, both of which are responsible for inflammatory responses and intestinal damage. Accordingly, BGUS activity has recently been proposed as a predictive marker for irinotecan-induced diarrhea [82], and it has been reported that the co-administration of irinotecan with a selective inhibitor of bacterial β-glucuronidases can prevent irinotecan toxicity in mice [83].

2. Conclusions and Future Perspectives

As mentioned above, the microbiota-based approach is an important resource in the therapy of PDAC, as it influences the efficacy and tolerability of chemotherapy, immunotherapy, and radiotherapy. Accordingly, the modulation of the microbiota is one of the most studied topics at present. These strategies include the use of FMT, probiotics, prebiotics, and antibiotics. Riquelme et al. studied the role of FMT in the mouse models of pancreatic cancer. They showed a better clinical response at five weeks in mice that received stools from patients who had the disease more than five years after resection compared to mice that had advanced PDAC patients as donors. In addition, their study showed a higher infiltration of CD8+ T cells into tumor tissue in mice that received FMT from patients who had the disease more than five years after resection, demonstrating the role of their microbiome in modulating immune response and survival [40]. FMT may also have an important effect in reducing the toxicity of chemotherapy, as demonstrated in animal models, suggesting a role in improving treatment adherence and compliance [81]. Interestingly, this effect is also observed with radiotherapy. In mice, the recovery rate after radiation treatment was shorter in animals receiving FMT, likely due to the differential expression of long noncoding RNA by microorganisms [84].
Probiotics and molecules derived from them may also be effective against pancreatic cancer and significantly reduce infectious complications after pancreatoduodenectomy, according to a recent study [85]. Ferrichrome, a substance produced by Lactobacillus casei ATCC334, can inhibit pancreatic, gastric, and colon cancer cells [52,53]. Ferrichrome could induce the M1 phenotype of tumor-associated macrophages (TAMs) via a TLR4-dependent pathway, thereby increasing immune system activities and the efficacy of immune checkpoint blocker therapies [86,87,88]. The administration of next-generation probiotics, such as Akkermansia muciniphila, Prevotella copri, Parabacteroides goldsteinii, and Faecalibacterium prausnitzii, represents a novel strategy [89]. In particular, A. muciniphila, a Gram-negative bacterium that can support gut immunity and cytokine release, significantly inhibits the proliferation of rat pancreatic islet tumor cells [90]. Interestingly, in certain neoplasms, such as renal or pulmonary carcinoma, the presence of resident microbiota has proven necessary for the effectiveness of immunotherapy. In a different way, in pancreatic carcinoma, the presence of certain bacteria appears to be a factor against therapeutic effectiveness. Indeed, the study of the mechanisms of chemical resistance induced by bacteria led to the recognition of specific targets to antagonize this effect. This includes, for example, CDA inhibitors and inhibitors of the bacterial enzyme β-glucuronidase (GUS), the clinical value of which has to be determined in the future.
The use of antibiotics in PDAC, either in combination with chemotherapeutics or immunotherapeutics, is currently being tested or appears to shorten overall survival [91]. Another goal of antibiotic administration and microbiota modulation is to improve the tolerability of therapy. An example is the improvement of irinotecan-induced diarrhea and the demonstration of reduced intestinal toxicity in germ-free mice or mice treated with broad-spectrum antibiotics.
On the other hand, it should be kept in mind that prolonged antibiotic therapies may lead to side effects, alter the balance between the host and microbiota in all body regions, and result in the selection of antibiotic-resistant bacterial species, so further studies are essential to evaluate the safety of such strategies. One area of great interest is the genetic manipulation of microbes with the aim of producing vectors that are capable of enhancing immune responses, spreading toxins, transferring DNA or RNA to specific targets, and altering the expression of oncogenes in cancer cells [92].
The main studies concern Salmonella typhimurium and Listeria monocytogenes, but human trials of this approach have yet to demonstrate superiority over conventional treatment. This requires more trials to better assess the clinical implication of this mechanism.
Finally, lyophilized probiotics and vancomycin may increase the efficacy of pembrolizumab [93] and nivolumab [94], although clinical trials have not yet been completed [95]. In summary, future perspectives are increasingly focused on clarifying the role of the microbiota in tumorigenesis and therapeutic resistance processes [96,97,98]. Combining microbiota modulation with conventional treatments such as chemotherapy, radiation, or immunotherapy seems to improve efficacy and tolerability and represents an important way forward. However, further studies specifically tailored to pancreatic cancer and performed in human models are needed for proper understanding and future clinical applications of this knowledge.

3. Materials and Methods

A review of the literature was performed to focus on the role of gut and tissue microbiota in influencing the effectiveness and metabolism of medical treatment for pancreatic cancer. The electronic databases that were searched included PubMed, Medline and Google Scholar, Scopus, and Embase. We focused on the following keywords and terms: “Pancreatic Cancer”, “Resistance”, Treatment”, “Therapy” AND “Microbiota”. We sought these terms within titles, abstracts, and keywords.
The studies included in this review were carefully reviewed by 2 authors. We included all papers with the full text available, original works, and metanalysis.
Exclusion criteria were language other than English and availability only of the abstracts.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics. CA: Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef]
  2. Laurent, L.; Vullierme, M.; Rebours, V.; Maire, F.; Hentic, O.; Francoz, C.; Durand, F.; Ruszniewski, P.; Lévy, P. Estimation of the prevalence of intraductal papillary mucinous neoplasm of the pancreas in the French population through patients waiting for liver transplantation. United Eur. Gastroenterol. J. 2017, 5, 499–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Mizrahi, J.D.; Surana, R.; Valle, J.W.; Shroff, R.T. Pancreatic cancer. Lancet 2020, 395, 2008–2020. [Google Scholar] [CrossRef] [PubMed]
  4. Ben, Q.-W.; Liu, J.; Sun, Y.-W.; Wang, L.-F.; Zou, D.-W.; Yuan, Y.-Z. Cigarette Smoking and Mortality in Patients with Pan-creatic Cancer: A Systematic Review and Meta-Analysis. Pancreas 2019, 48, 985–995. [Google Scholar] [CrossRef] [PubMed]
  5. Rawla, P.; Sunkara, T.; Gaduputi, V. Epidemiology of Pancreatic Cancer: Global Trends, Etiology and Risk Factors. World J. Oncol. 2019, 10, 10–27. [Google Scholar] [CrossRef]
  6. 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]
  7. Zhang, J.-J.; Jia, J.-P.; Shao, Q.; Wang, Y.-K. Diabetes mellitus and risk of pancreatic cancer in China: A meta-analysis based on 26 case-control studies. Prim. Care Diabetes 2019, 13, 276–282. [Google Scholar] [CrossRef]
  8. Gandhi, S.; de la Fuente, J.; Murad, M.H.; Majumder, S. Chronic Pancreatitis Is a Risk Factor for Pancreatic Cancer, and Incidence Increases with Duration of Disease: A Systematic Review and Meta-analysis. Clin. Transl. Gastroenterol. 2022, 13, e00463. [Google Scholar] [CrossRef]
  9. Bae, J.-M.; Shim, S.R. Coffee Consumption and Pancreatic Cancer Risk: A Meta-Epidemiological Study of Population-based Cohort Studies. Asian Pac. J. Cancer Prev. 2020, 21, 2793–2798. [Google Scholar] [CrossRef] [PubMed]
  10. Ilic, M.; Milicic, B.; Ilic, I. Association between oral contraceptive use and pancreatic cancer risk: A systematic review and meta-analysis. World J. Gastroenterol. 2021, 27, 2643–2656. [Google Scholar] [CrossRef] [PubMed]
  11. Laoveeravat, P.; Thavaraputta, S.; Vutthikraivit, W.; Suchartlikitwong, S.; Mingbunjerdsuk, T.; Motes, A.; Nugent, K.; Rakvit, A.; Islam, E.; Islam, S. Proton Pump Inhibitors and Histamine-2 Receptor Antagonists on the Risk of Pancreatic Cancer: A Systematic Review and Meta-analysis. J. Assoc. Physicians 2020, 113, 100–107. [Google Scholar] [CrossRef] [PubMed]
  12. Park, W.; Chawla, A.; O’Reilly, E.M. Pancreatic Cancer. A Review. J. Am. Med. Assoc. 2021, 326, 851–862. [Google Scholar] [CrossRef] [PubMed]
  13. Sohal, D.P.; Mangu, P.B.; Khorana, A.; Shah, M.A.; Philip, P.A.; O’Reilly, E.M.; Uronis, H.E.; Ramanathan, R.K.; Crane, C.H.; Engebretson, A.; et al. Metastatic Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2016, 34, 2784–2796. [Google Scholar] [CrossRef] [PubMed]
  14. Elsayed, M.; Abdelrahim, M. The Latest Advancement in Pancreatic Ductal Adenocarcinoma Therapy: A Review Article for the Latest Guidelines and Novel Therapies. Biomedicines 2021, 9, 389. [Google Scholar] [CrossRef]
  15. Khorana, A.; Mangu, P.B.; Berlin, J.; Engebretson, A.; Hong, T.S.; Maitra, A.; Mohile, S.G.; Mumber, M.; Schulick, R.; Shapiro, M.; et al. Potentially Curable Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2016, 34, 2541–2556. [Google Scholar] [CrossRef]
  16. 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]
  17. Tas, F.; Sen, F.; Keskin, S.; Kilic, L.; Yildiz, I. Prognostic factors in metastatic pancreatic cancer: Older patients are associated with reduced overall survival. Mol. Clin. Oncol. 2013, 1, 788–792. [Google Scholar] [CrossRef] [Green Version]
  18. 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] [Green Version]
  19. Oh, J.-K.; Weiderpass, E. Infection and Cancer: Global Distribution and Burden of Diseases. Ann. Glob. Health 2014, 80, 384–392. [Google Scholar] [CrossRef]
  20. Sarhadi, V.; Mathew, B.; Kokkola, A.; Karla, T.; Tikkanen, M.; Rautelin, H.; Lahti, L.; Puolakkainen, P.; Knuutila, S. Gut microbiota of patients with different subtypes of gastric cancer and gastrointestinal stromal tumors. Gut Pathog. 2021, 13, 11. [Google Scholar] [CrossRef]
  21. Schepis, T.; De Lucia, S.S.; Nista, E.C.; Manilla, V.; Pignataro, G.; Ojetti, V.; Piccioni, A.; Gasbarrini, A.; Franceschi, F.; Candelli, M. Microbiota in Pancreatic Diseases: A Review of the Literature. J. Clin. Med. 2021, 10, 5920. [Google Scholar] [CrossRef] [PubMed]
  22. Ahuja, M.; Schwartz, D.; Tandon, M.; Son, A.; Zeng, M.; Swaim, W.; Eckhaus, M.; Hoffman, V.; Cui, Y.; Xiao, B.; et al. Orai1-Mediated Antimicrobial Secretion from Pancreatic Acini Shapes the Gut Microbiome and Regulates Gut Innate Immunity. Cell Metab. 2017, 25, 635–646. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Mitsuhashi, K.; Nosho, K.; Sukawa, Y.; Matsunaga, Y.; Ito, M.; Kurihara, H.; Kanno, S.; Igarashi, H.; Naito, T.; Adachi, Y.; et al. Association of Fusobacteriums pecies in pancreatic cancer tissues with molecular features and prognosis. Oncotarget 2015, 6, 7209–7220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Torres, P.J.; Fletcher, E.M.; Gibbons, S.M.; Bouvet, M.; Doran, K.S.; Kelley, S.T. Characterization of the salivary microbiome in patients with pancreatic cancer. Peerj 2015, 3, e1373. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Lu, H.; Ren, Z.; Li, A.; Li, J.; Xu, S.; Zhang, H.; Jiang, J.; Yang, J.; Luo, Q.; Zhou, K.; et al. Tongue coating microbiome data distinguish patients with pancreatic head cancer from healthy controls. J. Oral Microbiol. 2019, 11, 1563409. [Google Scholar] [CrossRef] [PubMed]
  26. Shirazi, M.S.R.; Al-Alo, K.Z.K.; Al-Yasiri, M.H.; Lateef, Z.M.; Ghasemian, A. Microbiome Dysbiosis and Predominant Bacterial Species as Human Cancer Biomarkers. J. Gastrointest. Cancer 2020, 51, 725–728. [Google Scholar] [CrossRef] [PubMed]
  27. Öğrendik, M. Periodontal Pathogens in the Etiology of Pancreatic Cancer. Gastrointest. Tumors 2017, 3, 125–127. [Google Scholar] [CrossRef] [Green Version]
  28. Gnanasekaran, J.; Gallimidi, A.B.; Saba, E.; Pandi, K.; Berchoer, L.E.; Hermano, E.; Angabo, S.; Makkawi, H.; Khashan, A.; Daoud, A.; et al. Intracellular Porphyromonas gingivalis Promotes the Tumorigenic Behavior of Pancreatic Carcinoma Cells. Cancers 2020, 12, 2331. [Google Scholar] [CrossRef]
  29. 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] [Green Version]
  30. Guo, W.; Zhang, Y.; Guo, S.; Mei, Z.; Liao, H.; Dong, H.; Wu, K.; Ye, H.; Zhang, Y.; Zhu, Y.; et al. Tumor microbiome contributes to an aggressive phenotype in the basal-like subtype of pancreatic cancer. Commun. Biol. 2021, 4, 1019. [Google Scholar] [CrossRef]
  31. Del Castillo, E.; Meier, R.; Chung, M.; Koestler, D.C.; Chen, T.; Paster, B.J.; Charpentier, K.P.; Kelsey, K.T.; Izard, J.; Michaud, D.S. The Microbiomes of Pancreatic and Duodenum Tissue Overlap and Are Highly Subject Specific but Differ between Pan-creatic Cancer and Noncancer Subjects. Cancer Epidemiol. Biomark. Prev. 2019, 28, 370–383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Nejman, D.; Livyatan, I.; Fuks, G.; Gavert, N.; Zwang, Y.; Geller, L.T.; Rotter-Maskowitz, A.; Weiser, R.; Mallel, G.; Gigi, E.; et al. The human tumor microbiome is composed of tumor type–specific intracellular bacteria. Science 2020, 368, 973–980. [Google Scholar] [CrossRef] [PubMed]
  33. Gaiser, R.A.; Halimi, A.; Alkharaan, H.; Lu, L.; Davanian, H.; Healy, K.; Hugerth, L.W.; Ateeb, Z.; Valente, R.; Moro, C.F.; et al. Enrichment of oral microbiota in early cystic precursors to invasive pancreatic cancer. Gut 2019, 68, 2186–2194. [Google Scholar] [CrossRef] [Green Version]
  34. Ren, Z.; Jiang, J.; Xie, H.; Li, A.; Lu, H.; Xu, S.; Zhou, L.; Zhang, H.; Cui, G.; Chen, X.; et al. Gut microbial profile analysis by MiSeq sequencing of pancreatic carcinoma patients in China. Oncotarget 2017, 8, 95176–95191. [Google Scholar] [CrossRef] [Green Version]
  35. Candelli, M.; Franza, L.; Pignataro, G.; Ojetti, V.; Covino, M.; Piccioni, A.; Gasbarrini, A.; Franceschi, F. Interaction between Lipopolysaccharide and Gut Microbiota in Inflammatory Bowel Diseases. Int. J. Mol. Sci. 2021, 22, 6242. [Google Scholar] [CrossRef] [PubMed]
  36. Kadosh, E.; Snir-Alkalay, I.; Venkatachalam, A.; May, S.; Lasry, A.; Elyada, E.; Zinger, A.; Shaham, M.; Vaalani, G.; Mernberger, M.; et al. The gut microbiome switches mutant p53 from tumour-suppressive to oncogenic. Nature 2020, 586, 133–138. [Google Scholar] [CrossRef]
  37. Casero, R.A., Jr.; Stewart, T.M.; Pegg, A.E. Polyamine metabolism and cancer: Treatments, challenges and opportunities. Nat. Rev. Cancer 2018, 18, 681–695. [Google Scholar] [CrossRef]
  38. Alonso-Curbelo, D.; Ho, Y.-J.; Burdziak, C.; Maag, J.L.V.; Iv, J.P.M.; Chandwani, R.; Chen, H.-A.; Tsanov, K.M.; Barriga, F.M.; Luan, W.; et al. A gene–environment-induced epigenetic program initiates tumorigenesis. Nature 2021, 590, 642–648. [Google Scholar] [CrossRef]
  39. 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]
  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. Alam, A.; Levanduski, E.; Denz, P.; Villavicencio, H.S.; Bhatta, M.; Alhorebi, L.; Zhang, Y.; Gomez, E.C.; Morreale, B.; Senchanthisai, S.; et al. Fungal mycobiome drives IL-33 secretion and type 2 immunity in pancreatic cancer. Cancer Cell 2022, 40, 153–167.e11. [Google Scholar] [CrossRef] [PubMed]
  42. Chakladar, J.; Kuo, S.Z.; Castaneda, G.; Li, W.T.; Gnanasekar, A.; Yu, M.A.; Chang, E.Y.; Wang, X.Q.; Ongkeko, W.M. The Pancreatic Microbiome Is Associated with Carcinogenesis and Worse Prognosis in Males and Smokers. Cancers 2020, 12, 2672. [Google Scholar] [CrossRef] [PubMed]
  43. Kartal, E.; Schmidt, T.S.B.; Molina-Montes, E.; Rodríguez-Perales, S.; Wirbel, J.; Maistrenko, O.M.; Akanni, W.; Alhamwe, B.A.; Alves, R.J.; Carrato, A.; et al. A faecal microbiota signature with high specificity for pancreatic cancer. Gut 2022, 71, 1359–1372. [Google Scholar] [CrossRef] [PubMed]
  44. Fu, Y.; Ricciardiello, F.; Yang, G.; Qiu, J.; Huang, H.; Xiao, J.; Cao, Z.; Zhao, F.; Liu, Y.; Luo, W.; et al. The Role of Mitochondria in the Chemoresistance of Pancreatic Cancer Cells. Cells 2021, 10, 497. [Google Scholar] [CrossRef] [PubMed]
  45. Huang, X.; Li, M.; Hou, S.; Tian, B. Role of the microbiome in systemic therapy for pancreatic ductal adenocarcinoma (Review). Int. J. Oncol. 2021, 59, 101. [Google Scholar] [CrossRef]
  46. Lehouritis, P.; Cummins, J.; Stanton, M.; Murphy, C.T.; McCarthy, F.; 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] [Green Version]
  47. Voorde, J.V.; Sabuncuoğlu, S.; Noppen, S.; Hofer, A.; Ranjbarian, F.; Fieuws, S.; Balzarini, J.; Liekens, S. Nucleoside-catabolizing Enzymes in Mycoplasma-infected Tumor Cell Cultures Compromise the Cytostatic Activity of the Anticancer Drug Gemcitabine. J. Biol. Chem. 2014, 289, 13054–13065. [Google Scholar] [CrossRef] [Green Version]
  48. Tijeras-Raballand, A.; Hilmi, M.; Astorgues-Xerri, L.; Nicolle, R.; Bièche, I.; Neuzillet, C. Microbiome and pancreatic ductal adenocarcinoma. Clin. Res. Hepatol. Gastroenterol. 2021, 45, 101589. [Google Scholar] [CrossRef]
  49. 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. 2021, 108, 709–716. [Google Scholar] [CrossRef]
  50. Kesh, K.; Mendez, R.; Abdelrahman, L.; Banerjee, S.; Banerjee, S. Type 2 diabetes induced microbiome dysbiosis is associated with therapy resistance in pancreatic adenocarcinoma. Microb. Cell Factories 2020, 19, 75. [Google Scholar] [CrossRef]
  51. Arsenijevic, T.; Nicolle, R.; Bouchart, C.; D’Haene, N.; Demetter, P.; Puleo, F.; Van Laethem, J.-L. Pancreatic Cancer Meets Human Microbiota: Close Encounters of the Third Kind. Cancers 2021, 13, 1231. [Google Scholar] [CrossRef] [PubMed]
  52. Bronckaers, A.; Balzarini, J.; Liekens, S. The cytostatic activity of pyrimidine nucleosides is strongly modulated by Mycoplasma hyorhinis infection: Implications for cancer therapy. Biochem. Pharmacol. 2008, 76, 188–197. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, S.; Yang, Y.; Weng, W.; Guo, B.; Cai, G.; Ma, Y.; Cai, S. Fusobacterium nucleatum promotes chemoresistance to 5-fluorouracil by upregulation of BIRC3 expression in colorectal cancer. J. Exp. Clin. Cancer Res. 2019, 38, 14. [Google Scholar] [CrossRef] [PubMed]
  54. 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] [Green Version]
  55. Heshiki, Y.; Vazquez-Uribe, R.; Li, J.; Ni, Y.; Quainoo, S.; Imamovic, L.; Li, J.; Sørensen, M.; Chow, B.K.C.; Weiss, G.J.; et al. Predictable modulation of cancer treatment outcomes by the gut microbiota. Microbiome 2020, 8, 28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ruiz-Cordero, R.; Devine, W.P. Targeted Therapy and Checkpoint Immunotherapy in Lung Cancer. Surg. Pathol. Clin. 2020, 13, 17–33. [Google Scholar] [CrossRef] [PubMed]
  57. Cuevas, L.; Daud, A. Immunotherapy for melanoma. Semin. Cutan. Med. Surg. 2018, 37, 127–131. [Google Scholar] [CrossRef]
  58. Balachandran, V.P.; Beatty, G.L.; Dougan, S.K. Broadening the Impact of Immunotherapy to Pancreatic Cancer: Challenges and Opportunities. Gastroenterology 2019, 156, 2056–2072. [Google Scholar] [CrossRef]
  59. Feig, C.; Gopinathan, A.; Neesse, A.; Chan, D.S.; Cook, N.; Tuveson, D.A. The Pancreas Cancer Microenvironment. Clin. Cancer Res. 2012, 18, 4266–4276. [Google Scholar] [CrossRef] [Green Version]
  60. Elkrief, A.; Derosa, L.; Zitvogel, L.; Kroemer, G.; Routy, B. The intimate relationship between gut microbiota and cancer immunotherapy. Gut Microbes 2019, 10, 424–428. [Google Scholar] [CrossRef]
  61. Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.-L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef] [Green Version]
  62. 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]
  63. Dong, J.; Gao, H.-L.; Wang, W.-Q.; Yu, X.-J.; Liu, L. Bidirectional and dynamic interaction between the microbiota and therapeutic resistance in pancreatic cancer. Biochim. Biophys. Acta Rev. Cancer 2021, 1875, 188484. [Google Scholar] [CrossRef]
  64. Virtue, A.; Wang, H.; Yang, X.-F. MicroRNAs and Toll-like Receptor/Interleukin-1 Receptor Signaling. J. Hematol. Oncol. 2012, 5, 66. [Google Scholar] [CrossRef] [Green Version]
  65. Huang, Y.; Zhu, N.; Zheng, X.; Liu, Y.; Lu, H.; Yin, X.; Hao, H.; Tan, Y.; Wang, D.; Hu, H.; et al. Intratumor Microbiome Analysis Identifies Positive Association Between Megasphaera and Survival of Chinese Patients With Pancreatic Ductal Adenocarcinomas. Front. Immunol. 2022, 13, 785422. [Google Scholar] [CrossRef]
  66. Roy, S.; Trinchieri, S.R.G. Microbiota: A key orchestrator of cancer therapy. Nat. Rev. Cancer 2017, 17, 271–285. [Google Scholar] [CrossRef]
  67. Garajová, I.; Balsano, R.; Wang, H.; Leonardi, F.; Giovannetti, E.; Deng, D.; Peters, G.J. The role of the microbiome in drug resistance in gastrointestinal cancers. Expert Rev. Anticancer. Ther. 2021, 21, 165–176. [Google Scholar] [CrossRef]
  68. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved Survival with Ipilimumab in Patients with Metastatic Melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef]
  69. Baskar, R.; Dai, J.; Wenlong, N.; Yeo, R.; Yeoh, K.-W. Biological response of cancer cells to radiation treatment. Front. Mol. Biosci. 2014, 1, 24. [Google Scholar] [CrossRef] [Green Version]
  70. Li, Z.; Zhang, Y.; Hong, W.; Wang, B.; Chen, Y.; Yang, P.; Zhou, J.; Fan, J.; Zeng, Z.; Du, S. Gut microbiota modulate radiotherapy-associated antitumor immune responses against hepatocellular carcinoma Via STING signaling. Gut Microbes 2022, 14, 2119055. [Google Scholar] [CrossRef]
  71. Shen, S.; Lim, G.; You, Z.; Ding, W.; Huang, P.; Ran, C.; Doheny, J.; Caravan, P.; Tate, S.; Hu, K.; et al. Gut microbiota is critical for the induction of chemotherapy-induced pain. Nat. Neurosci. 2017, 20, 1213–1216. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Zhao, T.-S.; Xie, L.-W.; Cai, S.; Xu, J.-Y.; Zhou, H.; Tang, L.-F.; Yang, C.; Fang, S.; Li, M.; Tian, Y. Dysbiosis of Gut Microbiota Is Associated with the Progression of Radiation-Induced Intestinal Injury and Is Alleviated by Oral Compound Probiotics in Mouse Model. Front. Cell. Infect. Microbiol. 2021, 11, 717636. [Google Scholar] [CrossRef]
  73. Jian, Y.; Zhang, D.; Liu, M.; Wang, Y.; Xu, Z.-X. The Impact of Gut Microbiota on Radiation-Induced Enteritis. Front. Cell. Infect. Microbiol. 2021, 11, 586392. [Google Scholar] [CrossRef]
  74. 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] [PubMed]
  75. 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] [Green Version]
  76. García-González, A.P.; Ritter, A.D.; Shrestha, S.; Andersen, E.C.; Yilmaz, L.S.; Walhout, A.J. Bacterial Metabolism Affects the C. elegans Response to Cancer Chemotherapeutics. Cell 2017, 169, 431–441.e8. [Google Scholar] [CrossRef] [Green Version]
  77. Peretz, A.; Ben Shlomo, I.; Nitzan, O.; Bonavina, L.; Schaffer, P.M.; Schaffer, M. Clostridium difficile Infection: Associations with Chemotherapy, Radiation Therapy, and Targeting Therapy Treatments. Curr. Med. Chem. 2016, 23, 4442–4449. [Google Scholar] [CrossRef]
  78. Hato, S.V.; Khong, A.; de Vries, I.J.M.; Lesterhuis, W.J. Molecular Pathways: The Immunogenic Effects of Platinum-Based Chemotherapeutics. Clin. Cancer Res. 2014, 20, 2831–2837. [Google Scholar] [CrossRef] [Green Version]
  79. Yeung, C.; Chiau, J.C.; Cheng, M.; Chan, W.; Chang, S.; Chang, Y.; Jiang, C.; Lee, H. Modulations of probiotics on gut microbiota in a 5-fluorouracil-induced mouse model of mucositis. J. Gastroenterol. Hepatol. 2020, 35, 806–814. [Google Scholar] [CrossRef]
  80. Kato, S.; Hamouda, N.; Kano, Y.; Oikawa, Y.; Tanaka, Y.; Matsumoto, K.; Amagase, K.; Shimakawa, M. Probiotic Bifidobacterium bifidum G9-1 attenuates 5-fluorouracil-induced intestinal mucositis in mice via suppression of dysbiosis-related secondary inflammatory responses. Clin. Exp. Pharmacol. Physiol. 2017, 44, 1017–1025. [Google Scholar] [CrossRef] [PubMed]
  81. Chang, C.-W.; Lee, H.-C.; Li, L.-H.; Chiau, J.-S.C.; Wang, T.-E.; Chuang, W.-H.; Chen, M.-J.; Wang, H.-Y.; Shih, S.-C.; Liu, C.-Y.; et al. Fecal Microbiota Transplantation Prevents Intestinal Injury, Upregulation of Toll-Like Receptors, and 5-Fluorouracil/Oxaliplatin-Induced Toxicity in Colorectal Cancer. Int. J. Mol. Sci. 2020, 21, 386. [Google Scholar] [CrossRef] [Green Version]
  82. Chamseddine, A.N.; Ducreux, M.; Armand, J.-P.; Paoletti, X.; Satar, T.; Paci, A.; Mir, O. Intestinal bacterial β-glucuronidase as a possible predictive biomarker of irinotecan-induced diarrhea severity. Pharmacol. Ther. 2019, 199, 1–15. [Google Scholar] [CrossRef]
  83. Wallace, B.D.; Wang, H.; Lane, K.T.; Scott, J.E.; Orans, J.; Koo, J.S.; Venkatesh, M.; Jobin, C.; Yeh, L.-A.; Mani, S.; et al. Alleviating Cancer Drug Toxicity by Inhibiting a Bacterial Enzyme. Science 2010, 330, 831–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Cui, M.; Xiao, H.; Li, Y.; Zhou, L.; Zhao, S.; Luo, D.; Zheng, Q.; Dong, J.; Zhao, Y.; Zhang, X.; et al. Faecal microbiota transplantation protects against radiation-induced toxicity. EMBO Mol. Med. 2017, 9, 448–461. [Google Scholar] [CrossRef] [PubMed]
  85. Nomura, T.; Tsuchiya, Y.; Nashimoto, A.; Yabusaki, H.; Takii, Y.; Nakagawa, S.; Sato, N.; Kanbayashi, C.; Tanaka, O. Probi-otics reduce infectious complications after pancreaticoduodenectomy. Hepatogastroenterology 2007, 54, 661–663. [Google Scholar] [PubMed]
  86. Ijiri, M.; Fujiya, M.; Konishi, H.; Tanaka, H.; Ueno, N.; Kashima, S.; Moriichi, K.; Sasajima, J.; Ikuta, K.; Okumura, T. Ferri-chrome Identified from Lactobacillus Casei ATCC334 Induces Apoptosis through Its Iron-Binding Site in Gastric Cancer Cells. Tumor Biol. 2017, 39, 1010428317711311. [Google Scholar] [CrossRef] [Green Version]
  87. Konishi, H.; Fujiya, M.; Tanaka, H.; Ueno, N.; Moriichi, K.; Sasajima, J.; Ikuta, K.; Akutsu, H.; Tanabe, H.; Kohgo, Y. Probiotic-derived ferrichrome inhibits colon cancer progression via JNK-mediated apoptosis. Nat. Commun. 2016, 7, 12365. [Google Scholar] [CrossRef]
  88. Chaib, M.; Hafeez, B.B.; Mandil, H.; Daria, D.; Pingili, A.K.; Kumari, S.; Sikander, M.; Kashyap, V.K.; Chen, G.-Y.; Anning, E.; et al. Reprogramming of pancreatic adenocarcinoma immunosurveillance by a microbial probiotic siderophore. Commun. Biol. 2022, 5, 1181. [Google Scholar] [CrossRef]
  89. Chang, C.-J.; Lin, T.-L.; Tsai, Y.-L.; Wu, T.-R.; Lai, W.-F.; Lu, C.-C.; Lai, H.-C. Next generation probiotics in disease amelioration. J. Food Drug Anal. 2019, 27, 615–622. [Google Scholar] [CrossRef] [Green Version]
  90. Liu, M.-N.; Zhang, L.; Dong, X.-Y.; Liu, M.; Cheng, G.; Zhang, X.-L.; He, F.; Wang, G.-Q. Effects of Akkermansia muciniphila on the Proliferation, Apoptosis and Insulin Secretion of Rat Islet Cell Tumor Cells. Sichuan Da Xue Xue Bao Yi Xue Ban 2020, 51, 13–17. [Google Scholar]
  91. Mohindroo, C.; Rogers, J.E.; Hasanov, M.; Mizrahi, J.; Overman, M.J.; Varadhachary, G.R.; Wolff, R.A.; Javle, M.M.; Fogelman, D.R.; Pant, S.; et al. A retrospective analysis of antibiotics usage and effect on overall survival and progressive free survival in patients with metastatic pancreatic cancer. J. Clin. Oncol. 2019, 37, e15781. [Google Scholar] [CrossRef]
  92. Nguyen, V.H.; Min, J.-J. Salmonella-Mediated Cancer Therapy: Roles and Potential. Nucl. Med. Mol. Imaging 2017, 51, 118–126. [Google Scholar] [CrossRef]
  93. Live Biotherapeutic Product MRx0518 and Pembrolizumab Combination Study in Solid Tumors—Full Text View—Clinical-Trials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03637803 (accessed on 7 November 2022).
  94. Nivolumab (Anti-PD1), Tadalafil and Oral Vancomycin in People with Refractory Primary Hepatocellular Carcinoma or Liver Dominant Metastatic Cancer from Colorectal or Pancreatic Cancers—Full Text View—ClinicalTrials.Gov. Available online: https://clinicaltrials.gov/ct2/show/NCT03785210 (accessed on 9 November 2022).
  95. Amara, S.; Yang, L.V.; Tiriveedhi, V.; Muzaffar, M. Complex Role of Microbiome in Pancreatic Tumorigenesis: Potential Therapeutic Implications. Cells 2022, 11, 1900. [Google Scholar] [CrossRef] [PubMed]
  96. Le, D.T.; Picozzi, V.J.; Ko, A.H.; Wainberg, Z.A.; Kindler, H.; Wang-Gillam, A.; Oberstein, P.E.; Morse, M.A.; Zeh, H.J.; Weekes, C.D.; et al. Results from a Phase IIb, Randomized, Multicenter Study of GVAX Pancreas and CRS-207 Compared with Chemotherapy in Adults with Previously Treated Metastatic Pancreatic Adenocarcinoma (ECLIPSE Study). Clin. Cancer Res. 2019, 25, 5493–5502. [Google Scholar] [CrossRef] [PubMed]
  97. Frances, A.; Cordelier, P. The Emerging Role of Cytidine Deaminase in Human Diseases: A New Opportunity for Therapy? Mol. Ther. 2020, 28, 357–366. [Google Scholar] [CrossRef] [PubMed]
  98. Le, D.T.; Wang-Gillam, A.; Picozzi, V.; Greten, T.F.; Crocenzi, T.; Springett, G.; Morse, M.; Zeh, H.; Cohen, D.; Fine, R.L.; et al. Safety and Survival with GVAX Pancreas Prime and Listeria Monocytogenes–Expressing Mesothelin (CRS-207) Boost Vaccines for Metastatic Pancreatic Cancer. J. Clin. Oncol. 2015, 33, 1325–1333. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Possible effects of different bacterial strains and their metabolites on therapies of pancreatic cancer. Continuous lines indicate stimulation; dashed lines indicate inhibition.
Figure 1. Possible effects of different bacterial strains and their metabolites on therapies of pancreatic cancer. Continuous lines indicate stimulation; dashed lines indicate inhibition.
Biomedicines 11 00157 g001
Table 1. A panoramic of novel treatments in development for PC.
Table 1. A panoramic of novel treatments in development for PC.
Pathway InhibitorsTargeting DNA Damage Response (DDR) Targeting Immune SystemTargeting Tumor MetabolismTargeting Tumor Stroma Fibrosis, and Extracellular Matrix
KRASThe Poly (ADP-ribose) polymerase (PARP) InhibitorImmune Checkpoint Inhibitors (PD-L1/PD-1, CTLA-4)Targeting Tricarboxylic acid (TCA) enzymesTargeting Hyaluronic Acid
Neurotrophic Tyrosine Receptor Kinase (NTRK) fusion Vaccines Targeting Immune Cells/Signals inside the stroma
Neuregulin-1 gene (NRG1) Fusion CAR-t Cells Transfusion
CD-40, IL-10 (failed to provide benefit)
Table 2. Principal microbiological fundings in pancreatic cancer tissue.
Table 2. Principal microbiological fundings in pancreatic cancer tissue.
Tumour-Associated Microbiota in Pancreatic Cancer
Increased PresenceReduced Presence
PhylumEuryarchaeotaFirmicutes and Proteobacteria
Genus
-
Porphyromonas, Prevotella, Bifidobacterium and Synergistetes
-
Acinetobacter, Sphingopyxis and Pseudomonas (KRAS mutations)
-
Malassezia and Alternaria (Mycobiome)
-
Lactobacillus
-
Pseudoxanthomonas, Saccharopolyspora, Streptomyces (worse prognosis)
SpeciesFusobacterium nucleatum, Beutenbergia cavernae DSM 12333, Mycoplasma hypopneumoniae, Tolypothrix sp. PCC 7601, Acidovorax ebreuus TPSY, Agrobacterium radiobacter K84, Aggregatibacter aphorophilus NJ8700, Shigella sonnei Ss046, Salmonella enterica, Citrobacter freundii
-
Saccharomonospora Viridis DSM 43017
-
Alkalihalobacillus clausii (worse prognosis)
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

Nista, E.C.; Del Gaudio, A.; Del Vecchio, L.E.; Mezza, T.; Pignataro, G.; Piccioni, A.; Gasbarrini, A.; Franceschi, F.; Candelli, M. Pancreatic Cancer Resistance to Treatment: The Role of Microbiota. Biomedicines 2023, 11, 157. https://doi.org/10.3390/biomedicines11010157

AMA Style

Nista EC, Del Gaudio A, Del Vecchio LE, Mezza T, Pignataro G, Piccioni A, Gasbarrini A, Franceschi F, Candelli M. Pancreatic Cancer Resistance to Treatment: The Role of Microbiota. Biomedicines. 2023; 11(1):157. https://doi.org/10.3390/biomedicines11010157

Chicago/Turabian Style

Nista, Enrico Celestino, Angelo Del Gaudio, Livio Enrico Del Vecchio, Teresa Mezza, Giulia Pignataro, Andrea Piccioni, Antonio Gasbarrini, Francesco Franceschi, and Marcello Candelli. 2023. "Pancreatic Cancer Resistance to Treatment: The Role of Microbiota" Biomedicines 11, no. 1: 157. https://doi.org/10.3390/biomedicines11010157

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