This web page was created programmatically, to learn the article in its unique location you’ll be able to go to the hyperlink bellow:
https://www.nature.com/articles/s41575-025-01119-y
and if you wish to take away this text from our website please contact us
Russell, W. An handle on a attribute organism of most cancers. Br. Med. J. 2, 1356–1360 (1890).
Whipps, J. M., Lewis, Ok. & Cooke, R. C. in Fungi in Biological Control Systems (ed. Burge, M. N.) 161–187 (Manchester Univ. Press, 1988).
Huang, D. W., Sherman, B. T. & Lempicki, R. A. Bioinformatics enrichment instruments: paths towards the excellent practical evaluation of enormous gene lists. Nucleic Acids Res. 37, 1–13 (2009).
Di Bella, J. M., Bao, Y., Gloor, G. B., Burton, J. P. & Reid, G. High throughput sequencing strategies and evaluation for microbiome analysis. J. Microbiol. Methods 95, 401–414 (2013).
Staley, C. & Sadowsky, M. J. Practical issues for sampling and information evaluation in up to date metagenomics-based environmental research. J. Microbiol. Methods 154, 14–18 (2018).
Thomas, R. M. & Jobin, C. The microbiome and most cancers: is the “oncobiome” mirage actual? Trends Cancer 1, 24–35 (2015).
Rose, C., Parker, A., Jefferson, B. & Cartmell, E. The characterization of feces and urine: a assessment of the literature to tell superior therapy know-how. Crit. Rev. Environ. Sci. Technol. 45, 1827–1879 (2015).
Borges-Canha, M. et al. Role of colonic microbiota in colorectal carcinogenesis: a scientific assessment. Rev. Esp. Enferm. Dig. 107, 659–671 (2015).
Sobhani, I. et al. Microbial dysbiosis in colorectal most cancers (CRC) sufferers. PLoS ONE 6, e16393 (2011).
Schwabe, R. F. & Jobin, C. The microbiome and most cancers. Nat. Rev. Cancer 13, 800–812 (2013).
Rahib, L. et al. Projecting most cancers incidence and deaths to 2030: the sudden burden of thyroid, liver, and pancreas cancers within the United States. Cancer Res. 74, 2913–2921 (2014).
Siegel, R. L., Giaquinto, A. N. & Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 74, 12–49 (2024).
Cruz, M. S., Tintelnot, J. & Gagliani, N. Roles of microbiota in pancreatic most cancers growth and therapy. Gut Microbes 16, 2320280 (2024).
Da Silver Xavier, G. The cells of the islets of langerhans. J. Clin. Med. 7, 54 (2018).
Sedlack, A. J. H. et al. Update within the administration of gastroenteropancreatic neuroendocrine tumors. Cancer 130, 3090–3105 (2024).
Zhang, C.-Y. et al. Investigating the causal relationship between intestine microbiota and gastroenteropancreatic neuroendocrine neoplasms: a bidirectional Mendelian randomization research. Front. Microbiol. 15, 1420167 (2024).
Kurki, M. I. et al. FinnGen supplies genetic insights from a well-phenotyped remoted inhabitants. Nature 613, 508–518 (2023).
Kurilshikov, A. et al. Large-scale affiliation analyses establish host elements influencing human intestine microbiome composition. Nat. Genet. 53, 156–165 (2021).
Chen, Z., Wang, Z., Bao, H. & Ma, S. Gut microbiota and pancreatic most cancers threat, and the mediating position of immune cells and inflammatory cytokines: a Mendelian randomization research. Front. Immunol. 15, 1408770 (2024).
Dorrestein, P. C., Mazmanian, S. Ok. & Knight, R. Finding the lacking hyperlinks amongst metabolites, microbes, and the host. Immunity 40, 824–832 (2014).
Nejman, D. et al. The human tumor microbiome consists of tumor type-specific intracellular micro organism. Science 368, 973–980 (2020). A complete profiling of the intratumoural microbiome of seven totally different tumour varieties, together with PDAC, demonstrates the compositional variations between tumour microbiomes and that almost all micro organism are current intracellularly, particularly inside tumour and immune cells.
Massironi, S. et al. Intratumor microbiome in neuroendocrine neoplasms: a brand new associate of tumor microenvironment? A pilot research. Cells 11, 692 (2022).
Underhill, D. M. & Iliev, I. D. The mycobiota: interactions between commensal fungi and the host immune system. Nat. Rev. Immunol. 14, 405–416 (2014).
Honkanen, J. et al. Fungal dysbiosis and intestinal irritation in youngsters with beta-cell autoimmunity. Front. Immunol. 11, 468 (2020).
Salamon, D. et al. Analysis of the intestine mycobiome in grownup sufferers with sort 1 and sort 2 diabetes utilizing next-generation sequencing (NGS) with elevated sensitivity-pilot research. Nutrients 13, 1066 (2021).
Steenblock, C. et al. Viral infiltration of pancreatic islets in sufferers with COVID-19. Nat. Commun. 12, 3534 (2021).
Dotta, F. et al. Coxsackie B4 virus an infection of β cells and pure killer cell insulitis in recent-onset sort 1 diabetic sufferers. Proc. Natl Acad. Sci. USA 104, 5115–5120 (2007).
Müller, J. A. et al. SARS-CoV-2 infects and replicates in cells of the human endocrine and exocrine pancreas. Nat. Metab. 3, 149–165 (2021).
Isaacs, S. R. et al. Enteroviruses and threat of islet autoimmunity or sort 1 diabetes: systematic assessment and meta-analysis of managed observational research detecting viral nucleic acids and proteins. Lancet Diabetes Endocrinol. 11, 578–592 (2023).
Hruban, R. H., Goggins, M., Parsons, J. & Kern, S. E. Progression mannequin for pancreatic most cancers. Clin. Cancer Res. 6, 2969–2972 (2000). Comprehensive assessment illustrating the mannequin of development of pancreatic most cancers that’s needed for a basic understanding of this illness.
Iacobuzio-Donahue, C. A., Velculescu, V. E., Wolfgang, C. L. & Hruban, R. H. Genetic foundation of pancreas most cancers growth and development: insights from whole-exome and whole-genome sequencing. Clin. Cancer Res. 18, 4257–4265 (2012).
Rawla, P., Sunkara, T. & Gaduputi, V. Epidemiology of pancreatic most cancers: world traits, etiology and threat elements. World J. Oncol. 10, 10–27 (2019).
Jandhyala, S. M. et al. Altered intestinal microbiota in sufferers with persistent pancreatitis: implications in diabetes and metabolic abnormalities. Sci. Rep. 7, 43640 (2017).
Isaiah, A., Parambeth, J. C., Steiner, J. M., Lidbury, J. A. & Suchodolski, J. S. The fecal microbiome of canines with exocrine pancreatic insufficiency. Anaerobe 45, 50–58 (2017).
Zhang, X. M. et al. Intestinal microbial neighborhood differs between acute pancreatitis sufferers and wholesome volunteers. Biomed. Environ. Sci. 31, 81–86 (2018).
Hong, J. et al. Gut microbiome modifications related to persistent pancreatitis and pancreatic most cancers: a scientific assessment and meta-analysis. Int. J. Surg. 110, 5781–5794 (2024).
Sonika, U. et al. Mechanism of elevated intestinal permeability in acute pancreatitis: alteration in tight junction proteins. J. Clin. Gastroenterol. 51, 461–466 (2017).
Ammori, B. J. et al. Early enhance in intestinal permeability in sufferers with extreme acute pancreatitis: correlation with endotoxemia, organ failure, and mortality. J. Gastrointest. Surg. 3, 252–262 (1999).
Dougherty, M. W. & Jobin, C. Intestinal micro organism and colorectal most cancers: etiology and therapy. Gut Microbes 15, 2185028 (2023).
Thomas, R. M. Microbial molecules, metabolites, and malignancy. Neoplasia 60, 101128 (2025).
Matthaei, H., Schulick, R. D., Hruban, R. H. & Maitra, A. Cystic precursors to invasive pancreatic most cancers. Nat. Rev. Gastroenterol. Hepatol. 8, 141–150 (2011).
Basturk, O. et al. A revised classification system and proposals from the Baltimore consensus assembly for neoplastic precursor lesions within the pancreas. Am. J. Surg. Pathol. 39, 1730–1741 (2015).
Maker, A. V. et al. Cyst fluid biosignature to foretell intraductal papillary mucinous neoplasms of the pancreas with excessive malignant potential. J. Am. Coll. Surg. 228, 721–729 (2019).
Matthaei, H. et al. miRNA biomarkers in cyst fluid increase the prognosis and administration of pancreatic cysts. Clin. Cancer Res. 18, 4713–4724 (2012).
Pust, M.-M. et al. Absence of a pancreatic microbiome in intraductal papillary mucinous neoplasm. Gut 73, 1131–1141 (2024).
Caporaso, J. G. et al. Global patterns of 16S rRNA variety at a depth of tens of millions of sequences per pattern. Proc. Natl Acad. Sci. USA 108, 4516–4522 (2011).
Goodrich, J. Ok. et al. Conducting a microbiome research. Cell 158, 250–262 (2014). A vital manuscript outlining the mandatory steps to design, perform and analyse a microbiome research, no matter speculation or illness sort.
Li, S. et al. Pancreatic cyst fluid harbors a novel microbiome. Microbiome 5, 147 (2017).
Gaiser, R. A. et al. Enrichment of oral microbiota in early cystic precursors to invasive pancreatic most cancers. Gut 68, 2186–2194 (2019).
Wang, B. et al. The roles and interactions of Porphyromonas gingivalis and fusobacterium nucleatum in oral and gastrointestinal carcinogenesis: a story assessment. Pathogens 13, 93 (2024).
Pushalkar, S. et al. The pancreatic most cancers microbiome promotes oncogenesis by induction of innate and adaptive immune suppression. Cancer Discov. 8, 403–416 (2018).
Thomas, R. M. et al. Intestinal microbiota enhances pancreatic carcinogenesis in preclinical fashions. Carcinogenesis 39, 1068–1078 (2018).
Halimi, A. et al. Isolation of pancreatic microbiota from cystic precursors of pancreatic most cancers with intracellular progress and DNA damaging properties. Gut Microbes 13, 1983101 (2021).
Hingorani, S. R. et al. Preinvasive and invasive ductal pancreatic most cancers and its early detection within the mouse. Cancer Cell 4, 437–450 (2003).
Hingorani, S. R. et al. Trp53R172H and KrasG12D cooperate to advertise chromosomal instability and extensively metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 7, 469–483 (2005).
Hill, R. et al. PTEN loss accelerates KrasG12D-induced pancreatic most cancers growth. Cancer Res. 70, 7114–7124 (2010).
Saba, E. et al. Oral micro organism speed up pancreatic most cancers growth in mice. Gut 73, 770–786 (2024).
Ma, H., Luo, W. & Gu, Y. Does oral microbiota have an in depth relationship with pancreatic most cancers? a scientific assessment and meta-analysis. Ann. Surg. Oncol. 30, 8635–8641 (2023).
Storz, P. Acinar cell plasticity and growth of pancreatic ductal adenocarcinoma. Nat. Rev. Gastroenterol. Hepatol. 14, 296–304 (2017).
Kleeff, J. et al. Pancreatic most cancers. Nat. Rev. Dis. Primers 2, 16022 (2016).
McGuigan, A. et al. Pancreatic most cancers: a assessment of medical prognosis, epidemiology, therapy and outcomes. World J. Gastroenterol. 24, 4846–4861 (2018).
Raderer, M. et al. Association between Helicobacter pylori an infection and pancreatic most cancers. Oncology 55, 16–19 (1998).
Stolzenberg-Solomon, R. Z. et al. Helicobacter pylori seropositivity as a threat issue for pancreatic most cancers. J. Natl Cancer Inst. 93, 937–941 (2001).
Yu, G. et al. Seropositivity to Helicobacter pylori and threat of pancreatic most cancers. Cancer Epidemiol. Biomark. Prev. 22, 2416–2419 (2013).
Maisonneuve, P., Amar, S. & Lowenfels, A. B. Periodontal illness, edentulism, and pancreatic most cancers: a meta-analysis. Ann. Oncol. 28, 985–995 (2017).
Cutler, C. W., Kalmar, J. R. & Genco, C. A. Pathogenic methods of the oral anaerobe, Porphyromonas gingivalis. Trends Microbiol. 3, 45–51 (1995).
Ahn, J., Segers, S. & Hayes, R. B. Periodontal illness, Porphyromonas gingivalis serum antibody ranges and orodigestive most cancers mortality. Carcinogenesis 33, 1055–1058 (2012).
Michaud, D. S. et al. Plasma antibodies to oral micro organism and threat of pancreatic most cancers in a big European potential cohort research. Gut 62, 1764–1770 (2013).
Duan, D. et al. Advances in multi-omics built-in evaluation strategies primarily based on the intestine microbiome and their purposes. Front. Microbiol. 15, 1509117 (2024).
Farrell, J. J. et al. Variations of oral microbiota are related to pancreatic ailments together with pancreatic most cancers. Gut 61, 582–588 (2012). One of the earliest investigations demonstrating an affiliation of a microbiota, on this case the salivary microbiota, with pancreatic most cancers.
Ren, Z. et al. Gut microbial profile evaluation by MiSeq sequencing of pancreatic carcinoma sufferers in China. Oncotarget 8, 95176–95191 (2017).
Chen, J., Zhao, Ok.-N. & Vitetta, L. Effects of intestinal microbial-elaborated butyrate on oncogenic signaling pathways. Nutrients 11, 1026 (2019).
Encarnação, J. C. et al. Butyrate, a dietary fiber spinoff that improves irinotecan impact in colon most cancers cells. J. Nutr. Biochem. 56, 183–192 (2018).
Zhu, X. et al. Microbial metabolite butyrate promotes anti-PD-1 antitumor efficacy by modulating T cell receptor signaling of cytotoxic CD8 T cell. Gut Microbes 15, 2249143 (2023).
Turner, B. G. et al. Diagnosis of pancreatic neoplasia with EUS and FNA: a report of accuracy. Gastrointest. Endosc. 71, 91–98 (2010).
Chen, G., Liu, S., Zhao, Y., Dai, M. & Zhang, T. Diagnostic accuracy of endoscopic ultrasound-guided fine-needle aspiration for pancreatic most cancers: a meta-analysis. Pancreatology 13, 298–304 (2013).
Yao, D.-W., Qin, M.-Z., Jiang, H.-X. & Qin, S.-Y. Comparison of EUS−FNA and EUS−FNB for prognosis of strong pancreatic mass lesions: a meta-analysis of potential research. Scand. J. Gastroenterol. 59, 972–979 (2024).
Geller, L. T. et al. Potential position of intratumor micro organism in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 357, 1156–1160 (2017). This investigation demonstrates that resistance to the frequent pancreatic most cancers chemotherapy gemcitabine could be secondary to the presence of intratumoural Gammaproteobacteria that possesses the extra lively type (‘long form’) of the bacterial enzyme cytidine deaminase.
Thomas, R. M. & Jobin, C. Microbiota in pancreatic well being and illness: the subsequent frontier in microbiome analysis. Nat. Rev. Gastroenterol. Hepatol. 17, 53–64 (2020).
Kohi, S. et al. Alterations within the duodenal fluid microbiome of sufferers with pancreatic most cancers. Clin. Gastroenterol. Hepatol. 20, e196–e227 (2022).
Jackson, M. A. et al. Proton pump inhibitors alter the composition of the intestine microbiota. Gut 65, 749–756 (2016).
Aykut, B. et al. The fungal mycobiome promotes pancreatic oncogenesis by way of activation of MBL. Nature 574, 264–267 (2019).
Narunsky-Haziza, L. et al. Pan-cancer analyses reveal cancer-type-specific fungal ecologies and bacteriome interactions. Cell 185, 3789–3806.e17 (2022).
Alam, A. et al. Fungal mycobiome drives IL-33 secretion and sort 2 immunity in pancreatic most cancers. Cancer Cell 40, 153–167.e11 (2022).
Fletcher, A. A., Kelly, M. S., Eckhoff, A. M. & Allen, P. J. Revisiting the intrinsic mycobiome in pancreatic most cancers. Nature 620, E1–E6 (2023).
Xu, F., Saxena, D., Pushalkar, S. & Miller, G. Reply to: revisiting the intrinsic mycobiome in pancreatic most cancers. Nature 620, E7–E9 (2023).
Kartal, E. et al. A faecal microbiota signature with excessive specificity for pancreatic most cancers. Gut 71, 1359–1372 (2022). This research demonstrates the feasibility of faecal microbiota testing to detect pancreatic most cancers and, when the microbiome signature was mixed with the at the moment out there pancreatic most cancers tumour marker CA19-9, a fair increased diagnostic accuracy was reported.
Zhang, P. et al. Metagenomic evaluation reveals altered intestine virome and diagnostic potential in pancreatic most cancers. J. Med. Virol. 96, e29809 (2024).
Atarashi, Ok. et al. Induction of colonic regulatory T cells by indigenous Clostridium species. Science 331, 337–341 (2011).
Nutsch, Ok. M. & Hsieh, C.-S. T cell tolerance and immunity to commensal micro organism. Curr. Opin. Immunol. 24, 385–391 (2012).
Zhu, Y.-H. et al. Immunosuppression, immune escape, and immunotherapy in pancreatic most cancers: centered on the tumor microenvironment. Cell Oncol. 46, 17–48 (2023).
Ho, W. J., Jaffee, E. M. & Zheng, L. The tumour microenvironment in pancreatic most cancers — medical challenges and alternatives. Nat. Rev. Clin. Oncol. 17, 527–540 (2020).
Sethi, V. et al. Gut microbiota promotes tumor progress in mice by modulating immune response. Gastroenterology 155, 33–37.e6 (2018).
Yu, Q. et al. Intestinal microbiota modulates pancreatic carcinogenesis by intratumoral pure killer cells. Gut Microbes 14, 2112881 (2022).
Riquelme, E. et al. Tumor microbiome variety and composition affect pancreatic most cancers outcomes. Cell 178, 795–806.e12 (2019). Although pancreatic most cancers survival is historically low, there are sufferers who’re long-term survivors. This research studies that the tumour microbiome composition of short-term survivors is statistically totally different than long-term survivors by decreased α-diversity. This distinction influences the host immune response and possibly survival.
Turner, M. W. The position of mannose-binding lectin in well being and illness. Mol. Immunol. 40, 423–429 (2003).
Garred, P., Larsen, F., Seyfarth, J., Fujita, R. & Madsen, H. O. Mannose-binding lectin and its genetic variants. Genes Immun. 7, 85–94 (2006).
Afshar-Kharghan, V. The position of the complement system in most cancers. J. Clin. Invest. 127, 780–789 (2017).
Ikebe, M. et al. Lipopolysaccharide (LPS) will increase the invasive skill of pancreatic most cancers cells by the TLR4/MyD88 signaling pathway. J. Surg. Oncol. 100, 725–731 (2009).
Dai, Z.-L., Wu, G. & Zhu, W.-Y. Amino acid metabolism in intestinal micro organism: hyperlinks between intestine ecology and host well being. Front. Biosci. 16, 1768–1786 (2011).
Lin, R., Liu, W., Piao, M. & Zhu, H. A assessment of the connection between the intestine microbiota and amino acid metabolism. Amino Acids 49, 2083–2090 (2017).
al-Waiz, M., Mikov, M., Mitchell, S. C. & Smith, R. L. The exogenous origin of trimethylamine within the mouse. Metab. Clin. Exp. 41, 135–136 (1992).
Mirji, G. et al. The microbiome-derived metabolite TMAO drives immune activation and boosts responses to immune checkpoint blockade in pancreatic most cancers. Sci. Immunol. 7, eabn0704 (2022).
Heath-Pagliuso, S. et al. Activation of the Ah receptor by tryptophan and tryptophan metabolites. Biochemistry 37, 11508–11515 (1998).
Shinde, R. & McGaha, T. L. The aryl hydrocarbon receptor: connecting immunity to the microenvironment. Trends Immunol. 39, 1005–1020 (2018).
Xue, P., Fu, J. & Zhou, Y. The aryl hydrocarbon receptor and tumor immunity. Front. Immunol. 9, 286 (2018).
Hezaveh, Ok. et al. Tryptophan-derived microbial metabolites activate the aryl hydrocarbon receptor in tumor-associated macrophages to suppress anti-tumor immunity. Immunity 55, 324–340.e8 (2022).
Roager, H. M. & Licht, T. R. Microbial tryptophan catabolites in well being and illness. Nat. Commun. 9, 3294 (2018).
Agrawal, R. & Natarajan, Ok. N. Oncogenic signaling pathways in pancreatic ductal adenocarcinoma. Adv. Cancer Res. 159, 251–283 (2023).
Yang, Q. et al. A assessment of intestine microbiota-derived metabolites in tumor development and most cancers remedy. Adv. Sci. 10, e2207366 (2023).
D’Amico, D. et al. Impact of the pure compound urolithin A on well being, illness, and ageing. Trends Mol. Med. 27, 687–699 (2021).
Totiger, T. M. et al. Urolithin A, a novel pure compound to focus on PI3K/AKT/mTOR pathway in pancreatic most cancers. Mol. Cancer Ther. 18, 301–311 (2019).
Morrison, D. J. & Preston, T. Formation of quick chain fatty acids by the intestine microbiota and their affect on human metabolism. Gut Microbes 7, 189–200 (2016).
Corrêa-Oliveira, R., Fachi, J. L., Vieira, A., Sato, F. T. & Vinolo, M. A. R. Regulation of immune cell perform by short-chain fatty acids. Clin. Transl. Immunol. 5, e73 (2016).
Bachem, A. et al. Microbiota-derived short-chain fatty acids promote the reminiscence potential of antigen-activated CD8+ T cells. Immunity 51, 285–297.e5 (2019).
Coutza, C. et al. Systemic quick chain fatty acids restrict antitumor impact of CTLA-4 blockade in hosts with most cancers. Nat. Commun. 11, 2165 (2020).
Nomura, M. et al. Association of short-chain fatty acids within the intestine microbiome with medical response to therapy with nivolumab or pembrolizumab in sufferers with strong most cancers tumors. JAMA Netw. Open 3, e202895 (2020).
Ziemons, J. et al. Fecal ranges of SCFA and BCFA throughout capecitabine in sufferers with metastatic or unresectable colorectal most cancers. Clin. Exp. Med. 23, 3919–3933 (2023).
Sono, M. et al. Reduction of butyrate-producing micro organism within the intestine microbiome of Japanese sufferers with pancreatic most cancers. Pancreatology 24, 1031–1039 (2024).
Zhou, W. et al. The fecal microbiota of sufferers with pancreatic ductal adenocarcinoma and autoimmune pancreatitis characterised by metagenomic sequencing. J. Transl. Med. 19, 215 (2021).
Weir, T. L. et al. Stool microbiome and metabolome variations between colorectal most cancers sufferers and wholesome adults. PLoS ONE 8, e70803 (2013).
Wu, N. et al. Dysbiosis signature of fecal microbiota in colorectal most cancers sufferers. Microb. Ecol. 66, 462–470 (2013).
Mirzaei, R. et al. Role of microbiota-derived short-chain fatty acids in most cancers growth and prevention. Biomed. Pharmacother. 139, 111619 (2021).
Markowiak-Kopeć, P. & Śliżewska, Ok. The impact of probiotics on the manufacturing of short-chain fatty acids by human intestinal microbiome. Nutrients 12, 1107 (2020).
Yang, X. et al. Clostridium butyricum and its metabolite butyrate promote ferroptosis susceptibility in pancreatic ductal adenocarcinoma. Cell Oncol. 46, 1645–1658 (2023).
Irajizad, E. et al. A blood-based metabolomic signature predictive of threat for pancreatic most cancers. Cell Rep. Med. 4, 101194 (2023).
Nagata, N. et al. Metagenomic identification of microbial signatures predicting pancreatic most cancers from a multinational research. Gastroenterology 163, 222–238 (2022).
Wallace, B. D. et al. Alleviating most cancers drug toxicity by inhibiting a bacterial enzyme. Science 330, 831–835 (2010).
de Castilhos, J. et al. Microbiome and pancreatic most cancers: time to consider chemotherapy. Gut Microbes 16, 2374596 (2024).
Yang, W. et al. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 manufacturing and intestine immunity. Nat. Commun. 11, 4457 (2020).
Ratajczak, W. et al. Immunomodulatory potential of intestine microbiome-derived short-chain fatty acids (SCFAs). Acta Biochim. Pol. 66, 1–12 (2019).
Tintelnot, J. et al. Microbiota-derived 3-IAA influences chemotherapy efficacy in pancreatic most cancers. Nature 615, 168–174 (2023).
Fu, S.-F. et al. Indole-3-acetic acid: a widespread physiological code in interactions of fungi with different organisms. Plant Signal. Behav. 10, e1048052 (2015).
Von Hoff, D. D. et al. Increased survival in pancreatic most cancers with nab-paclitaxel plus gemcitabine. N. Engl. J. Med. 369, 1691–1703 (2013).
Conroy, T. et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic most cancers. N. Engl. J. Med. 364, 1817–1825 (2011).
Shiravand, Y. et al. Immune checkpoint inhibitors in most cancers remedy. Curr. Oncol. 29, 3044–3060 (2022).
Vitiello, G. A., Cohen, D. J. & Miller, G. Harnessing the microbiome for pancreatic most cancers immunotherapy. Trends Cancer 5, 670–676 (2019).
Sethi, V., Vitiello, G. A., Saxena, D., Miller, G. & Dudeja, V. The position of the microbiome in immunologic growth and its implication for pancreatic most cancers immunotherapy. Gastroenterology 156, 2097–2115.e2 (2019).
Rogers, S., Charles, A. & Thomas, R. M. The prospect of harnessing the microbiome to enhance immunotherapeutic response in pancreatic most cancers. Cancers 15, 5708 (2023).
Han, Z.-Y. et al. Probiotics functionalized with a gallium-polyphenol community modulate the intratumor microbiota and promote anti-tumor immune responses in pancreatic most cancers. Nat. Commun. 15, 7096 (2024).
Ni, Y. et al. Distinct composition and metabolic features of human intestine microbiota are related to cachexia in lung most cancers sufferers. ISME J. 15, 3207–3220 (2021).
Yang-Jensen, S. Ok. et al. Intestinal host-microbe interactions gasoline pulmonary irritation in cigarette smoke uncovered mice. Gut Microbes 17, 2519699 (2025).
Dilmore, A. H. et al. Medication use is related to distinct microbial options in nervousness and melancholy. Mol. Pyschiatry 30, 2545–2557 (2025).
Szajewska, H. et al. Antibiotic-perturbed microbiota and the position of probiotics. Nat. Rev. Gastroenterol. Hepatol. 22, 155–172 (2025).
Mirzayi, C. et al. Reporting tips for human microbiome analysis: the STORMS guidelines. Nat. Med. 27, 1885–1892 (2021).
Knight, R. et al. Best practices for analysing microbiomes. Nat. Rev. Microbiol. 16, 410–422 (2018).
Fierer, N. et al. Guidelines for stopping and reporting contamination in low-biomass microbiome research. Nat. Microbiol. 10, 1570–1580 (2025).
Salter, S. J. et al. Reagent and laboratory contamination can critically affect sequence-based microbiome analyses. BMC Biol. 12, 87 (2014).
Caruso, V., Song, X., Asquith, M. & Karstens, L. Performance of microbiome sequence inference strategies in environments with various biomass. mSystems 4, e00163-18 (2019).
Eisenhofer, R. et al. Contamination in low microbial biomass microbiome research: points and proposals. Trends Microbiol. 27, 105–117 (2019).
He, Y. et al. Regional variation limits purposes of wholesome intestine microbiome reference ranges and illness fashions. Nat. Med. 24, 1532–1535 (2018).
Olsson, L. M. et al. Dynamics of the conventional intestine microbiota: a longitudinal one-year inhabitants research in Sweden. Cell Host Microbe 30, 726–739.e3 (2022).
Xu, Z., Malmer, D., Langille, M. G. I., Way, S. F. & Knight, R. Which is extra vital for classifying microbial communities: who’s there or what they’ll do? ISME J. 8, 2357–2359 (2014).
Abbas, S. et al. Bacteriophage remedy: a doable various remedy towards antibiotic-resistant strains of Klebsiella pneumoniae. Front. Microbiol. 16, 1443430 (2025).
Yarahmadi, A. et al. Beyond antibiotics: exploring multifaceted approaches to fight bacterial resistance within the trendy period: a complete assessment. Front. Cell. Infect. Microbiol. 15, 1493915 (2025).
Li, Y., Li, X.-M., Duan, H.-Y., Yang, Ok. & Ye, J.-F. Advances and optimization methods in bacteriophage remedy for treating inflammatory bowel illness. Front. Immunol. 15, 1398652 (2024).
Xiao, Y. et al. The position of bacteriophage in inflammatory bowel illness and its therapeutic potential. Crit. Rev. Microbiol. (2025).
Gilbert, J. A. et al. Clinical translation of microbiome analysis. Nat. Med. 31, 1099–1113 (2025).
Porcari, S. et al. International consensus assertion on microbiome testing in medical follow. Lancet Gastroenterol. Hepatol. 10, 154–167 (2025).
Tran, Ok. A. et al. Deep studying in most cancers prognosis, prognosis and therapy choice. Genome Med. 13, 152 (2021).
McCulloch, J. A. et al. Intestinal microbiota signatures of medical response and immune-related hostile occasions in melanoma sufferers handled with anti-PD-1. Nat. Med. 28, 545–556 (2022).
Wu, G. et al. A core microbiome signature as an indicator of well being. Cell 187, 6550–6565.e11 (2024).
Derosa, L. et al. Custom scoring primarily based on ecological topology of intestine microbiota related to most cancers immunotherapy consequence. Cell 187, 3373–3389.e16 (2024).
Lazaros, Ok. et al. Non-invasive biomarkers within the period of massive information and machine studying. Sensors 25, 1396 (2025).
Arulvasan, W. et al. Optimized breath evaluation: custom-made analytical strategies and enhanced workflow for broader detection of VOCs. Metabolomics 21, 17 (2025).
Hutkins, R. et al. Classifying compounds as prebiotics — scientific views and proposals. Nat. Rev. Gastroenterol. Hepatol. 22, 54–70 (2025).
Armengaud, J. The daybreak of the revolution that can permit us to exactly describe how microbiomes perform. J. Proteom. 316, 105430 (2025).
Zaramela, L. S. et al. The sum is larger than the elements: exploiting microbial communities to attain complicated features. Curr. Opin. Biotechnol. 67, 149–157 (2021).
Sharma, Ok. R., Colvis, C. M., Rodgers, G. P. & Sheeley, D. M. Illuminating the druggable genome: pathways to progress. Drug Discov. Today 29, 103805 (2024).
Villani, A. et al. A strong machine studying method to establish interactions of differentially plentiful intestine microbial subsets in sufferers with metastatic and non-metastatic pancreatic most cancers. Gut Microbes 16, 2375483 (2024).
Bolyen, E. et al. Reproducible, interactive, scalable and extensible microbiome information science utilizing QIIME 2. Nat. Biotechnol. 37, 852–857 (2019).
Shalon, D. et al. Profiling the human intestinal setting beneath physiological circumstances. Nature 617, 581–591 (2023).
Schmidt, F. et al. Noninvasive evaluation of intestine perform utilizing transcriptional recording sentinel cells. Science 376, eabm6038 (2022).
This web page was created programmatically, to learn the article in its unique location you’ll be able to go to the hyperlink bellow:
https://www.nature.com/articles/s41575-025-01119-y
and if you wish to take away this text from our website please contact us
This web page was created programmatically, to learn the article in its authentic location you…
This web page was created programmatically, to learn the article in its unique location you…
This web page was created programmatically, to learn the article in its unique location you…
This web page was created programmatically, to learn the article in its authentic location you…
This web page was created programmatically, to learn the article in its unique location you…
This web page was created programmatically, to learn the article in its authentic location you'll…