Graphical abstract graphic file with name fx1.jpg [63]Open in a new tab Highlights * • Cigarette smoke exposure (CSE) induces unique gut microbial and metabolomic signatures * • Gut microbial dysbiosis induced by CSE is essential for its tumor-promoting effects * • Fecal microbiome transfer from CSE mice into smoke-naïve mice drives increased tumor growth * • Targeting the dysbiotic gut microbiome ameliorates tumor-promoting effects of CSE __________________________________________________________________ Biological sciences; Immunology; Microbiome; Cancer; Metabolomics Introduction Cigarette smoke exposure (CSE) has been extensively investigated as a risk factor for cancer development.[64]^1^,[65]^2 Potential carcinogens detected in cigarette smoke[66]^3 have been postulated to aid cancer initiation and progression through genotoxic effects and reactive oxygen species generation.[67]^4^,[68]^5 Recent studies have also explored the direct effects on cell proliferation through binding to nicotinic acetylcholine receptors (nAChRs).[69]^6 However, the molecular mechanisms underlying the tumor-promoting effects of CSE remain elusive, and there is a dearth of clinically actionable targets. We have witnessed the recent emergence of the gut microbiome as a key regulator of human physiology, and its perturbations linked to ailments such as gastrointestinal diseases,[70]^7 neurological diseases,[71]^8 behavioral conditions,[72]^9 and malignancies.[73]^10^,[74]^11^,[75]^12^,[76]^13^,[77]^14^,[78]^15 Recent research has demonstrated that smokers, too, harbor a dysbiotic microbiome.[79]^16^,[80]^17^,[81]^18 However, the implications of this changed gut microbiota have not yet been explored. We, along with others, have shown that the gut microbiome significantly drives cancer progression by affecting the anti-tumor immune response.[82]^14^,[83]^19 Targeting the microbiome as a strategy to enhance the efficacy of immunotherapy in multiple cancers is being explored.[84]^20 In light of these findings, we hypothesized that the gut microbiome might be the missing link between smoking and cancer progression. Herein, we elucidate a novel mechanism for smoking-dependent cancer progression via the gut microbiome through modulation of the adaptive immune system. We characterize the unique microbiome and metabolome potentiating this smoking-dependent cancer progression and present an actionable target to mitigate the harmful effects of cigarette smoke. Results The gut microbiome is required and sufficient to mediate cigarette-smoke-induced cancer progression We first evaluated the effect of gut microbiome depletion on cigarette smoke exposure (CSE)-induced tumor progression. CSE was given to cancer-naive C57BL/6J wild-type (WT) mice with and without a broad-spectrum antibiotic cocktail to deplete the gut microbiome followed by subcutaneous cancer cell implantation ([85]Figure 1A). CSE promoted tumor growth in KPC (pancreatic cancer), MC-38 (colon cancer), and MB-49 (bladder cancer) models in the presence of gut microbiome ([86]Figures 1B–1D, left panels; [87]Figure S1A). Interestingly, in gut-microbiome-depleted states, CSE failed to promote tumor growth ([88]Figures 1B–1D, right panels). In another set of experiments, when the smoke analog nicotine-derived nitrosamine ketone (NNK; N-methyl-N-(4-oxo-4-pyridin-3-ylbutyl)nitrous amide), one of the most potent carcinogens in cigarette smoke,[89]^21 was administered intraperitoneally, gut microbiome depletion was still able to nullify any tumor-promoting effects across multiple cancer models ([90]Figures S1B–S1D). Figure 1. [91]Figure 1 [92]Open in a new tab The gut microbiome is required and sufficient to promote cigarette-smoke-induced cancer progression (Also refer to [93]Figure S1) (A) Schematic timeline of experimental design of the subcutaneous tumor model in C57BL/6J mice showing the pre-exposure phase of 4 weeks before tumor implantation followed by continued exposure of CSE with or without gut microbiome depletion. (B–D) Subcutaneous tumor volumes at the endpoint in mice exposed to CSE with or without gut microbiome depletion using (B) pancreatic cancer (data representative of three independent experiments), (C) colon cancer, and (D) bladder cancer cell lines, respectively (n = 8–12 per group). (Left panels (B–D): gut microbiome present; right panels (B–D): gut microbiome depleted state). (E) Pancreatic weights of LSL-Kras^G12D/+;Pdx-1-Cre (KC mice) at 4 months of age after exposure with NNK for 2 months with or without gut microbiome depletion. (n = 11–12 per group). (F) (Top panel) Representative H&E stain of KC mice pancreas showing significant ductal proliferation and fibrosis in the pancreas of mice exposed to NNK compared to controls, gut microbiome depleted, and gut microbiome depleted NNK-exposed mice (scale bar, 563.6 μm); (Middle panel) Trichrome staining showing fibrosis in the pancreas of KC mice. Quantification (shown on the left) done as described in methodology (n = 4 mice per group; scale bar, 563.6 μm); (bottom panel) cytokeratin-19 staining showing ductal proliferation among various groups. (n = 4 per group; scale bar, 281.8 μm). (G) Tumor kinetics of control and CSE mice with CSE mice implanted with Mc-38 cell line randomized at day 14 to receive no antibiotics vs. broad-spectrum antibiotics. (n = 10 controls: n = 20 smoke-exposed before randomization at day 14). (H) Schematic timeline of FMT studies in C57BL/6J mice randomized to receive FMT from control mice or CSE/NNK-exposed mice. (I) Subcutaneous tumor volumes at the endpoint in recipient mice getting FMT from control donors vs. CSE donors (n = 12 per group). Data representative of two independent experiments. CSE: cigarette smoke exposure; NNK: 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone; FMT: fecal microbiome transplant. ∗ p value <0.05; ∗∗p value <0.01; ∗∗∗ p value <0.001. Unpaired t test or ANOVA used as appropriate for statistical comparison. Data presented as mean ± SEM. Next, we utilized genetic models with a predisposition to cancer formation to test our hypothesis ([94]Figure S1E). NNK exposure to KC (LSL-Kras^G12D/+;Pdx-1-Cre) mice, which are predisposed to spontaneous pancreatic intraepithelial neoplasia (PanIN) and pancreatic cancer development, resulted in increased mean pancreatic weights compared to mice receiving simultaneous NNK and broad-spectrum antibiotic treatment ([95]Figure 1E). Histology showed significant fibrosis and increased CK-19 expression in the NNK-exposed group relative to the control group; however, simultaneous treatment with antibiotics to NNK-exposed mice significantly reduced fibrosis and ductal proliferation ([96]Figure 1F). Similarly, A/J mice, which are highly susceptible to forming carcinogen-induced lung tumors, developed significantly more tumor nodules in the lungs of NNK-exposed mice with intact gut microbiomes when compared to those with depleted gut microbiomes ([97]Figure S1F). To confirm whether this effect could be replicated in the setting of well-established tumors, we employed a therapeutic model of gut microbiome depletion where subcutaneous tumors were allowed to grow for 14 days under the influence of CSE before antibiotic depletion was initiated. The broad-spectrum antibiotic cocktail was able to significantly retard tumor growth in mice receiving CSE after only 5 days of treatment ([98]Figure 1G). To further evaluate if smoking-induced gut microbial dysbiosis is sufficient to cause smoke-induced tumor progression, we performed fecal microbiota transplants (FMTs) from tumor-naive control (smoke-free) WT donor mice and cigarette-smoke-exposed WT donor mice into recipient WT mice with depleted gut microbiome ([99]Figure 1H). These recipient mice with reconstituted gut microbiomes were then challenged with subcutaneous KPC pancreatic cancer tumors. Mice receiving FMT from smoke-exposed mice had significantly higher mean tumor volumes than mice receiving FMT from control mice ([100]Figure 1I). To further validate this finding, FMT was performed with NNK-exposed mice instead of CSE mice with similar results ([101]Figure S1G). We measured serum cotinine concentrations (a byproduct of nicotine metabolism, which has been shown to correlate with smoke exposure[102]^22) in the donor mice as well as recipient mice post-FMT to ensure that smoke metabolites were not being passively transferred through the FMT. Serum cotinine levels were significantly elevated only in the CSE-exposed donor mice (25.8 ng/mL), consistent with the levels reported in the literature for mice undergoing smoke exposure.[103]^5 Mice receiving FMT from smoke donors had relatively undetectable levels (<0.1 ng/mL), making the transmission of nicotine through donor stool unlikely ([104]Figure S1H). In addition, antibiotics also did not affect serum cotinine levels ([105]Figure S1I). A recent report has indicated that gut microbes can metabolize and degrade nicotine.[106]^23 To verify whether antibiotics treatment was affecting nicotine levels in the gut, we analyzed the nicotine levels in ileal contents. Although mice exposed to cigarette smoke had higher levels of nicotine in the ileal contents, these remained unchanged in mice where gut microbiome was sterilized with antibiotics, indicating that nicotine metabolism was not affected by gut microbiome in our model ([107]Figure S1J). These findings indicate that the gut microbiome is required and sufficient to induce smoking-dependent tumor progression. Modulation of adaptive and innate immune response drives the smoking-induced-dysbiosis-mediated cancer progression We next characterized the immune tumor microenvironment (TME) of subcutaneous tumors from control and smoke-exposed mice with or without gut microbiome depletion through flow cytometry. Smoke-exposed mice had a globally attenuated anti-tumor immune response with decreased CD3^+ lymphocytes, CD4^+ T cells, CD8^+ T cells, and CD11c+ MHC II + dendritic cells. On the other hand, tumor-promoting moieties, such as myeloid-derived suppressor cells (MDSCs [CD45^+ CD11b+ Ly6G+]), were upregulated. The M1 (CD45+F4/80+MHCII+CD206-) to M2 (CD45+F4/80+MHCII-CD206+) macrophage ratio was tilted significantly in favor of the M2 phenotype ([108]Figures 2A–2C and [109]S2A–S2C). Gut microbial depletion was able to rescue the anti-tumor immune machinery with downregulation of MDSCs and upregulation of antigen-presenting dendritic cells, tumor-infiltrating CD4^+ T cells, and CD8^+ T cells; however, the macrophage polarization could not be reversed ([110]Figure S2C). Intriguingly, this immunomodulation of the TME was also evident in mice receiving FMT from NNK-exposed donors. Immunophenotyping of tumors implanted in NNK FMT recipient mice revealed reduced infiltration of CD3^+ T cells, CD4^+ T cells, and CD8^+ T cells, along with significantly elevated MDSCs ([111]Figures 2D–2F and [112]S2D). Th17 T cells (CD3^+CD4+IL17+), known to potentiate an immunosuppressive phenotype in the tumor microenvironment, increased with NNK FMT but did not reach statistical significance ([113]Figure S2E). Moreover, tumor growth inhibition seen with therapeutic antibiotic administration to smoke-exposed mice was accompanied by significantly increased CD8^+ T cell infiltration. MDSCs (CD45^+CD11b+Ly6G+) infiltration in the TME simultaneously decreased; however, it did not reach statistical significance ([114]Figures S2F and S2G). These findings indicate that gut microbiome depletion rescues the anti-tumor adaptive immune machinery in the TME. Figure 2. [115]Figure 2 [116]Open in a new tab Modulation of adaptive immune response drives the smoking-induced dysbiosis-mediated cancer progression (Also refer to [117]Figures S2 and [118]S3) (A–C) Graphical representation of tumor flow-cytometric analysis (left) and bar graphs (right) of control, CSE, antibiotics alone, and CSE + antibiotics group showing percentage infiltration of CD3CD4+ T cells (A), CD3CD8+ T cells (B), and CD11b+Ly6G + MDSC (C) cells of single cells in the tumor microenvironment, respectively, showing CSE-induced immunosuppressive changes reversed with gut microbiome depletion (n = 7–10 per group). (D–F) Tumor flow-cytometric analysis showing immunosuppressive changes characterized by decreased CD3CD4+ T cells (D), CD3CD8+ T cells (E), and increased CD11b+Ly6G + MDSC cells (F), respectively, induced in mice that received FMT from NNK-exposed mice vs. controls (n = 7–8 per group). (G) Tumor volumes in WT and Rag1-KO mice exposed to CSE with or without gut microbiome depletion with broad-spectrum antibiotics (n = 7–10 per group). (H) Tumor volumes in WT and Rag1-KO mice receiving FMT from smoke donors (smoke FMT) vs. control donors (control FMT) (n = 9–12 for WT mice, n = 8–9 for Rag1-KO mice). (I) Tumor volumes in WT (n = 14–15) and CD8-KO mice (n = 7–8) exposed to CSE. (J) Tumor kinetics in WT CSE mice with or without Ly6G-depleting monoclonal antibody with appropriate controls (n = 10 per group). ∗p value <0.05; ∗∗p value <0.01; ∗∗∗p value <0.001. Unpaired t test or ANOVA used as appropriate for statistical comparison. Data presented as mean ± SEM. To confirm the importance of the adaptive immune system, we first utilized the Rag1-KO mouse model, which lacks a functional adaptive immune system.[119]^24 We found that CSE did not promote tumor growth in Rag1-KO mice; additionally, in these mice, gut microbiome depletion failed to decrease the tumor burden ([120]Figure 2G). Administration of NNK instead of CSE to Rag1-KO mice yielded similar results ([121]Figure S2H). This indicates that the adaptive immune system is essential for smoking-induced gut microbial dysbiosis to affect tumor progression. To eliminate the possibility that potential differences in the baseline microbiome of Rag1-KO mice, when compared to the WT mice, could be confounding our results, we reconstituted the gut-microbiome-depleted Rag1-KO mice with FMT from either smoke-exposed or smoke-free WT mice. The tumor-promoting effect of the smoke-exposed dysbiotic gut microbiome was abrogated in Rag1-KO mice ([122]Figure 2H). To specifically delineate the role of the CD8 adaptive immune arm, we evaluated the effect of smoking in CD8-KO mice and found CSE did not promote tumor growth, signifying that the effector immune cells are essential links for the dysbiotic microbiome to mediate its effects ([123]Figure 2I). To test whether antibiotics treatment led to increased cytotoxicity in CD8^+ T cells, we performed an ex vivo cytotoxicity study by incubating splenic CD8^+ T cells, isolated from tumor-bearing mice, with calcein-AM-labeled KPC cells (see [124]STAR Methods). CD8^+ T cells isolated from CSE mice treated with antibiotics had significantly greater cytotoxicity against KPC cells ex vivo, as compared to CSE mice without antibiotics ([125]Figure S3A). Additionally, Ki67 staining of tumors obtained from CSE + antibiotics mice demonstrated less cancer cell proliferation as compared to CSE mice ([126]Figure S3B), indicating that increased CD8^+ T cell infiltration and cytotoxicity may be leading to decreased cancer cell proliferation. As smoke-exposed tumors showed increased accumulation of CD11b+Ly6G + cells, which have been previously shown to decrease cytotoxic CD8^+ T cell infiltration in the TME,[127]^25 we hypothesized that these cells were responsible for decreased effector CD8 T cell tumor infiltration in smoke-exposed mice. Specifically targeting these cells using anti-Ly6G monoclonal antibody (Invivomab Catalog# BE0075-1, Clone 1A8), with cell depletion confirmed on flow cytometry of splenocytes ([128]Figure S3C), prevented the tumor-promoting effects of smoking ([129]Figure 2J). Smoke-induced dysbiosis is characterized by unique microbial and metabolomic signatures amenable to selective targeting Gut microbial profiling by 16S rRNA sequencing revealed that the CSE group clustered separately compared to the control group upon principal coordinate analysis (PCoA) ([130]Figure S4A). Linear regression analysis showed significant enrichment of gram-negative genera (Bacteroides and Akkermansia) and certain gram-positive genera (Turicibacter, Acetatifactor, and Blautia) in the CSE group ([131]Figure S4B). Similarly, NNK exposure induced a significant difference in beta diversity ([132]Figure S4C), and interestingly, gram-negative genera like Bacteroides and Akkermansia were enriched upon NNK exposure as well ([133]Figure S4D). When the gut microbiome composition of mice bearing subcutaneous tumors was evaluated, the CSE group again demonstrated a distinct gut microbial signature. In KPC subcutaneous tumor-bearing mice, bacteria belonging to Akkermansia spp., Turicibacter spp., Faecalibaculum spp., and Pseudomonas spp. were the most enriched in the CSE group ([134]Figure 3A). Furthermore, CSE led to a significantly different gut microbial composition in mice bearing subcutaneous MC-38 colon cancer ([135]Figure S4E) and MB-49 bladder tumors ([136]Figure S4G). Interestingly, despite a different tumor microenvironment, bacteria from genus Akkermansia were the enriched bacteria in the gut in the CSE group in both tumor types, similar to KPC pancreatic tumors ([137]Figures S3F and [138]S4H). Additionally, tumor-bearing mice that were receiving smoke FMT had significant enrichments of bacteria from the gram-negative genera Prevotella, Parabacteroides, and Alloprevotella compared to mice that received control FMT ([139]Figure 3B). Figure 3. [140]Figure 3 [141]Open in a new tab Smoke-induced dysbiosis is characterized by unique microbial and metabolomic signatures amenable to selective targeting (Also refer to [142]Figures S4 and [143]S5) (A) Fecal samples were obtained from subcutaneous KPC tumor-bearing control and CSE mice and analyzed using 16s rRNA amplicon sequencing for metagenomic characterization. (Left panel) The significantly enriched microbes based on linear model analysis in the CSE (green bars) and control (red bars) groups are shown. (Right panel) PCoA plot comparing beta diversity between control (red) and CSE (green) groups. Distance was calculated using Bray-Curtis analysis, and PERMANOVA was used for statistical significance. (B) Fecal samples obtained from C57BL/6J mice receiving FMT from control or CSE donors and implanted with KPC pancreatic tumors were similarly interrogated through 16 s rRNA sequencing. Linear model (left panel) and Bray-Curtis analysis (right panel) are shown. (C) Heatmap representation of all differential fecal metabolites (BH FDR <0.25) between tumor-bearing control (n = 10) and CSE (n = 11) mice. A total of 194 differential metabolites were obtained upon untargeted LC-MS analysis. (D) Significantly altered metabolic pathways were obtained using pathway enrichment analysis of significantly altered fecal metabolites upon CSE using MetaboAnalyst 5.0. Only the metabolites annotated in HMDB were used for the analysis (88/194 differential metabolites). (E) Subcutaneous tumor volumes at endpoint after CSE with selective targeting of gut microbiome using oral neomycin gavage (200 mg/kg) or complete gut microbiome depletion using broad-spectrum antibiotics cocktail (n = 8–12 per group). (F) Flow cytometric analysis of tumors showing % of CD3^+CD8^+ of single cells after selective targeting of gut microbiome using oral neomycin gavage (200 mg/kg) or broad-spectrum antibiotic cocktail (n = 7–8 per group). ∗p value <0.05, ∗∗p value <0.01. CSE, cigarette smoke exposure; PCoA, principle coordinate analysis; PERMANOVA, permutational analysis of variance; FMT, fecal microbiome transplant; LC-MS, liquid chromatography-mass spectrometry; BH, Benjamini-Hochberg; FDR, false discovery rate; HMDB, human metabolome database. Unpaired t test or ANOVA used, as appropriate, for statistical comparison. Data presented as mean ± SEM. To evaluate the gut microbial metabolomic signature induced by CSE, we performed untargeted liquid chromatography-mass spectrometry (LC-MS) metabolomics on fecal samples from tumor-bearing control and CSE mice. A total of 781 unique metabolites were identified. Unbiased hierarchical clustering using a scale-normalized concentration of discriminatory metabolites with a false discovery rate <0.25 in control and smoke-exposed groups showed that the two groups had distinct signatures, with 194 metabolites having a significantly differential abundance ([144]Table S1; [145]Figure 3C). Qualitative metabolic set enrichment analysis of metabolites that were significantly altered in the CSE group compared to the controls revealed that arginine biosynthesis and arachidonic acid metabolism were the top metabolic pathways that were altered due to CSE ([146]Figure 3D). Since CSE led to the enrichment of both gram-positive and gram-negative bacterial species, we utilized selective targeting of these microbes using narrow-spectrum antibiotics. Neomycin, which has a predominantly gram-negative spectrum, significantly reduced tumor burden in CSE mice. In contrast, vancomycin (exclusively gram-positive spectrum) showed only partial tumor reduction ([147]Figure S5A). Additionally, neomycin demonstrated an anti-tumor effect equivalent to the broad-spectrum antibiotic cocktail ([148]Figure 3E). Neomycin treatment partially ameliorated the metabolomic changes induced due to CSE ([149]Figures S5B and S5C). Finally, selective targeting with neomycin also reverses the immunosuppressive changes in the TME, as evidenced by increased CD8^+ T cell tumor infiltration ([150]Figure 3F). Discussion We report one of the first studies demonstrating the role of gut microbiome in propagating pro-tumorigenic effects of CSE across multiple smoking-dependent cancers. CSE was associated with an immunosuppressive TME, and its effect was neutralized when the gut microbiome was depleted. FMT experiments confirmed the tumor-promoting and immunosuppressive effects of this CSE-modulated gut microbiome. Our results are in line with a recent study by Bai et al., where cigarette smoke expedited colon cancer growth in an azoxymethane-induced colitis model through enrichment of Eggerthella lenta.[151]^26 Prior pre-clinical investigations have focused on alterations in cell signaling pathways, such as nicotinic receptor stimulation or genetic/epigenetic changes in proto-oncogenes and tumor suppressor genes, as mechanistic explanations for smoking-induced carcinogenesis. However, in our study, FMT experiments suggest that CSE-induced gut microbial dysbiosis can independently exert tumor-promoting effects. Moreover, ablation of the gut microbiome in mice receiving CSE led to abrogation of tumor growth, whereas the levels of smoke metabolites in serum (cotinine) or stool (polyaromatic hydrocarbons) were unaffected. This suggests that even in the presence of circulating CSE components, the gut microbiome plays an essential role in promoting tumor growth. Exposure to smoke, or FMT from CSE mice, led to significant upregulation of granulocytic MDSCs in the TME, and this immunosuppressive phenotype was reversed with gut microbiome depletion. In addition, MDSC-specific marker antibody depletion was able to reverse the tumor-promoting effects of smoking, implicating a possible mechanistic link between the dysbiotic gut microbiome induced by CSE and myeloid cell infiltration. Interestingly, Pushalkar et al. similarly demonstrated that the gut microbiome can modulate MDSC infiltration in the TME in KC genetic model of pancreatic cancer.[152]^19 Another dominant immunosuppressive population in pancreatic cancer TMEs is tumor-associated macrophages.[153]^27 Prior work from Kumar et al. indicates that smoke exposure can induce differentiation of Ly6G + MDSCs into tumor-associated macrophages (TAMs), which secrete epidermal growth factor receptor (EGFR) ligands to accelerate PanIN development in the KC mouse model.[154]^28 Similarly, we found increased M2-polarized macrophages (F4/80+, MHC II−, CD206 +) in the TME upon CSE; however, this could not be reversed upon gut microbial ablation, indicating that macrophage polarization likely does not explain the effects of gut microbial targeting on tumor growth. Additionally, CSE significantly impacted adaptive immune response in the TME and decreased CD8 T cell infiltration. These findings are concordant with recent human studies in esophageal, head, and neck squamous cell cancer, where smokers had decreased CD8 T cell infiltration in the TME.[155]^29^,[156]^30 Treatment with antibiotics was able to increase CD8^+ T cell infiltration in the TME as well as CD8^+ T cell cytotoxicity on ex vivo analysis. The loss of pro-tumorigenic effects of CSE in Rag1-KO and CD8-KO mice, along with the failure of smoke-FMT to increase tumor growth in Rag1-KO mice, suggests the need for functional adaptive immunity for CSE dysbiosis to promote tumor growth. Characterization of the CSE dysbiotic gut microbiome through 16s rRNA sequencing revealed tumor-specific enrichment patterns. However, certain microbes, such as Akkermansia spp. and the Clostridiales vadin BB60 group, were consistently elevated across tumor types. Akkermansia has been favorably associated with immunomodulatory responses in obesity and homeostatic immunity but also has been pathologically indicated in disease states like multiple sclerosis.[157]^31^,[158]^32^,[159]^33^,[160]^34 The gut microbiome is known to exert its effects in a context-specific manner.[161]^10 It is possible that under the influence of CSE, Akkermansia assumes a pro-tumorigenic phenotype. However, further experiments are needed to substantiate such conclusions. We did observe significant anti-tumor activity with neomycin treatment that might indicate a specific role for gram-negative dysbiosis in smoke-mediated tumor progression. There have been recent reports regarding accumulation of nicotine in the intestinal lumen and its degradation by gut bacteria in the context of NASH cirrhosis.[162]^23 Interestingly, in our study, although nicotine levels in the luminal contents of ileum increased with smoke exposure, we did not observe any differences in nicotine levels upon antibiotics treatment. This combined with unchanged serum cotinine levels in smoke-exposed mice upon antibiotics treatment may indicate that in our model, nicotine metabolism by gut bacterium did not play a significant role. This discrepancy may reflect the different biological context, when comparing NASH cirrhosis to implanted cancer models. However, further studies are needed to establish whether nicotine metabolism by gut microbiome is essential for tumor-promoting effects of cigarette smoke. CSE also led to alterations in composition of stool metabolites that are known to exert immunomodulatory effects. CSE was associated with significantly altered arachidonic acid metabolism, which was partially reversed through neomycin treatment. The arachidonic acid pathway can significantly affect the induction of inflammation in both benign and malignant disease states. Prostaglandin E2 can induce profound immunosuppression via various mechanisms, including infiltration of MDSCs in the TME,[163]^35 whereas leukotriene B4 is a potent myeloid chemoattractant.[164]^36^,[165]^37 We also observed significant alterations in the arginine biosynthesis pathway on CSE. L-arginine is essential for T lymphocytes to mount an anti-tumor immune response,[166]^38 and depletion of extracellular arginine by MDSC-expressed arginase I can lead to immunosuppression in the tumor microenvironment.[167]^39 Interestingly, our data indicate downregulation of L-arginine upon CSE and subsequent increase upon neomycin treatment. Bai et al.[168]^26 identified alterations in bile-acid metabolism that could explain the effects of smoking-induced dysbiosis on colon cancer progression. In their experiments, taurodeoxycholic acid (TCDA), a secondary bile acid, was upregulated upon smoke exposure, and the authors posited that this could promote cancer growth through the MAP/ERK signaling pathway. In our stool metabolomic profiling, we did not observe an enrichment of bile acid metabolism in CSE mice. The highly context-specific composition and phenotypic effects might explain the differences in the microbes and their metabolites identified in our studies. For example, in our study, antibiotics treatment alone decreased the tumor burden in the KPC model but did not affect the tumor burden in MC-38 or MB-49 models. Regardless, antibiotics treatment consistently decreased the tumor-promoting effects of CSE in all these models. In summary, we demonstrate the induction of a unique metabolic and microbial signature upon smoke exposure that creates an immunosuppressive tumor microenvironment and favors cancer progression. Limitations of the study Our study does have certain limitations. It is unclear how smoking-induced dysbiosis is established and what mechanisms underlie the interaction between the dysbiotic gut microbiome and anti-cancer immune response. Whether these effects are mediated by specific microbes or a group of microbes sharing a specific functional phenotype needs to be clarified. For example, we did notice a preponderance of gram-negative bacteria upon CSE; however, the mechanisms by which these bacteria interact with and modulate the innate and/or adaptive immune system has not been explored in our study. Additionally, the role of immunomodulatory metabolic pathways altered upon CSE needs to be investigated. Resource availability Lead contact Further information and requests for resources should be directed to and will be fulfilled by the lead contact, Vikas Dudeja (vdudeja@uabmc.edu). Materials availability No new unique reagents were generated in this study. Data and code availability * • Data: the metabolomics data raw files can be accessed through NIH metabolomics workbench using Data track ID 4229 and Study ID is ST002823. The 16 S rRNA raw sequencing files can be accessed using the NCBI Bio project accession number [169]PRJNA1007862. The following link can be used to access the data after the release date. [170]https://www.ncbi.nlm.nih.gov/sra/PRJNA1007862. * • Code: the code used for processing and analyzing the 16s rRNA data can be accessed at [171]https://github.com/asorgen/Smoking_Cancer_Gut_Dysbiosis_Analys is_2023.git. All analysis was done using R ([172]https://www.r-project.org/). * • All other data are available upon request from the [173]lead contact. Acknowledgments