key: cord-0028009-33jkkbft authors: Desai, Sanket; Dharavath, Bhasker; Manavalan, Sujith; Rane, Aishwarya; Redhu, Archana Kumari; Sunder, Roma; Butle, Ashwin; Mishra, Rohit; Joshi, Asim; Togar, Trupti; Apte, Shruti; Bala, Pratyusha; Chandrani, Pratik; Chopra, Supriya; Bashyam, Murali Dharan; Banerjee, Anirban; Prabhash, Kumar; Nair, Sudhir; Dutt, Amit title: Fusobacterium nucleatum is associated with inflammation and poor survival in early-stage HPV-negative tongue cancer date: 2022-03-04 journal: NAR Cancer DOI: 10.1093/narcan/zcac006 sha: aae5224289a539b1c9e8e48edcbbfbf072f11af8 doc_id: 28009 cord_uid: 33jkkbft Persistent pathogen infection is a known cause of malignancy, although with sparse systematic evaluation across tumor types. We present a comprehensive landscape of 1060 infectious pathogens across 239 whole exomes and 1168 transcriptomes of breast, lung, gallbladder, cervical, colorectal, and head and neck tumors. We identify known cancer-associated pathogens consistent with the literature. In addition, we identify a significant prevalence of Fusobacterium in head and neck tumors, comparable to colorectal tumors. The Fusobacterium-high subgroup of head and neck tumors occurs mutually exclusive to human papillomavirus, and is characterized by overexpression of miRNAs associated with inflammation, elevated innate immune cell fraction and nodal metastases. We validate the association of Fusobacterium with the inflammatory markers IL1B, IL6 and IL8, miRNAs hsa-mir-451a, hsa-mir-675 and hsa-mir-486-1, and MMP10 in the tongue tumor samples. A higher burden of Fusobacterium is also associated with poor survival, nodal metastases and extracapsular spread in tongue tumors defining a distinct subgroup of head and neck cancer. The Human Microbiome Project has identified 48 microbial habitats in the human body (1) . These microbes maintain balanced symbiotic/commensal relationships or a 'eubiosis' under normal conditions (2) . A shift in the eubiotic balance or a 'dysbiosis' can lead to disease. Chronic inflammation, often linked to cancer initiation and pro-gression, is known to result from these dysbiotic events and persistent infections of specific microbes. The infections also elicit host immune responses making the microenvironment tumor-permissive (3) . Additionally, genotoxins or processed metabolites from the microbes have also been shown to induce genomic instability and are known to modulate the tumorigenesis (4). Considering the above factors to define possible causality and formulation of models of cancer pathogenesis due to microbes (especially in colon cancer) provide a framework for studying the role of cancerassociated microbes across different tissues (5) . Few well-known associations between microbial infection and cancer are that of Helicobacter pylori, which is causally related to 60-90% of all gastric cancer cases (6) . Causality between infection and tumorigenesis has been established in various animal model systems for H. pylori (7, 8) . Other reported associations are those of Salmonella typhi infection with gallbladder cancer (9, 10) , Streptococcus bovis, enterotoxigenic Bacteroides fragilis, genotoxic Escherichia coli and Fusobacterium nucleatum with colon cancer (11- 15) , and Chlamydia pneumonia with lung cancer (16) . Interestingly, 20% of all human microbial habitats are associated with the head and neck region (17) . Multiple studies have investigated the enrichment of distinct microbial species in the head and neck squamous cell carcinomas (HNSCs). They have shown an association of microbial burden with the stage of the disease (18, 19) . Multiple studies suggest that the association between periodontal bacteria and tumorigenesis is mediated by chronic inflammation. Recently reported systematic meta-analyses highlight role of F. nucleatum in head and neck cancer (20) . Other inflammatory periodontal bacterial pathogens have also been implicated to be an independent risk factor in HPV (human papillomavirus)-negative HNSCs (21) . Currently, minimal information exists regarding the association between such periodontal pathogens and biology, especially in HPVnegative HNSC tumors. Systematic efforts are required to understand the role of specific intratumor microbes and their clinical implications. Next-generation sequencing (NGS) provides a powerful and unbiased tool for identification of viral and bacterial pathogens from human samples, including tumor microenvironment (22) . Microbial composition has been derived from human tissue using various computational approaches, including computational subtraction (23) . Building on this computational subtraction approach, we recently developed infectious pathogen detector (IPD) (24) , to identify infectious pathogens from heterogeneous NGS datasets. Using IPD, we performed systematic quantification of 1060 infectious pathogens from 239 DNA and 1168 RNA sequencing samples representing six cancer types: breast, cervical, colorectal, lung, gallbladder and HNSC. Along with identifying known tumor-associated microbes, we find enrichment of F. nucleatum in the oral tumors, consistent with earlier reports (25) . We observed mutual exclusivity of HPV and Fusobacterium in HNSCs, with both forming a distinct subclass of tumors. Comparing the gene and miRNA expression data within the HPV-negative HNSC tumors, we identified differentially expressed genes and miRNAs associated with a higher burden of Fusobac-terium. We further validated the selected inflammation markers and inflammation-linked miRNAs in the in-house tongue tumor patient samples. Correlating with the immune cell signatures, we identify that the Fusobacterium-high subgroup of HNSC tumors shows increased innate immunity factors, with pro-tumorigenic potential. Locoregional lymph node metastasis and tumor extension outside lymph node capsule are known poor prognosis factors in head and neck cancer (26) . This study defines a distinct subgroup of HPV-negative tongue tumors with higher inflammation, nodal metastases, extracapsular spread and poor prognosis, characterized by potential surrogate miRNA markers. The cervical adenocarcinoma patient samples (n = 41) were collected from cervical adenocarcinoma patients at the Tata Memorial Hospital (TMH) and ACTREC biorepository. The study was approved by the ACTREC-Tata Memorial Centre (TMC) institutional review board (IRB) (study protocol #116). From the 41 cervical samples, whole exome sequencing was performed on 17 paired tumor samples and 1 orphan tumor sample (n = 35), and whole transcriptome sequencing was performed on 24 tumor and 5 normal samples (n = 29). The retrospectively collected non-small cell lung cancer patient samples (n = 52) were obtained from the TMH and ACTREC biorepository. Whole exome sequencing was performed on all the samples (n = 52). Sample set and study protocols were approved by the IRB and Ethics Committee (EC) of TMC-ACTREC (study protocol # 900514). The retrospectively collected oral cancer samples were obtained from the TMH and ACTREC tumor tissue repository, with clinical characteristics as described in Table 1 . Fresh frozen primary tumor (n = 75) and normal (n = 25) oral samples were collected at the TMH and ACTREC, Mumbai, India. The sample set and study protocols were approved by the IRB and EC of TMC-ACTREC (study protocol #88). The anatomical sites of all the oral cancer samples collected were of tongue origin and were identified to be earlystage tumors by clinical assessment. Of the total 75 oral tumor samples, 1 sample was ignored due to low RNA concentration. The set of 74 tumor and 25 normal samples was used as a validation set to quantify F. nucleatum, human genes and miRNAs associated with the pathogen. The primer sequences for each microRNA, genes and Fusobacterium validation used in the study are given in Supplementary Table S1 . For exome sequencing of 35 cervical samples, two different capture kits were employed. In brief, SureSelect XT Target Enrichment Kit (Agilent Technologies, Santa Clara, CA, USA) capturing 50 Mb of the genome was used for 13 samples. Genomic DNA (200 ng) was sheared using covaris to generate 150-500 bp fragment size. For remaining to obtain an average coverage of 100× across all the samples. The exome sequencing method used for the 27 paired colorectal samples used in the study is described in (28) . For transcriptome sequencing of 29 cervical samples, library preparation was performed using SENSE Total RNA-Seq Library Prep Kit (Lexogen Inc.). The ribosomal RNA (rRNA) was depleted from total RNA using Ribo-Cop rRNA Depletion Kit V1.2 (Lexogen Inc.) as per the manufacturer's protocol. Libraries were sequenced in the Illumina HiSeq 4000 platform to yield 100 bp paired-end reads, with total output of >60 million reads per sample. For colorectal transcriptome samples, only tumors harboring >70% of tumor epithelial cells were used for RNA isolation. RNA was isolated from colorectal samples using Trizol reagent (Invitrogen Inc., Carlsbad, CA, USA) as per the manufacturer's protocol. Five micrograms of total RNA was used to prepare the RNA sequencing library using the TruSeq RNA Sample Prep Kits (Illumina). Removal of abundant rRNA from a total RNA sample was performed with the Epicentre Ribo-Zero family of rRNA depletion kits and libraries were prepared as per the manufacturer's protocol (Illumina). Generated libraries were sequenced on HiSeq 2000/2500 to obtain 2 × 100 bp paired-end reads to obtain 140 million clean reads. We developed IPD, a computational tool, to quantify pathogens from the whole exome and transcriptome samples, as described earlier (24) . IPD performs data preprocessing by filtering the low-quality and ambiguous alignment reads and gives normalized quantification of fragments aligned to pathogen genome, by scaling it to library yield [fragments per million (FPM)] and normalizing to genome length [fragments per kilobase per million (FPKM)]. These normalizations make the pathogen quantification comparable across different samples and across library types (single-and paired-end data). Samples having an FPM of >0 were considered as positive for the bacterial pathogen. Specifically for viral pathogens, a stringent threshold of 1 FPM was used for positivity, as described earlier (29) . In total, 239 exomes and 110 transcriptomes of inhouse samples representing six cancer types (breast, lung, gallbladder, cervical, colorectal and oral) were analyzed using IPD (details are provided in Supplementary Table S2 ). In addition, 1058 TCGA RNA-seq sample data consisting of 512 (472 tumors, 40 normal) colon adenocarcinomas (TCGA-COAD) and 546 (502 tumor, 44 normal) head and neck cancers (TCGA-HNSC) were also analyzed using IPD. Sample-wise IPD-based normalized pathogen quantification for these samples was compiled using in-house Python scripts. Heatmap representations were generated using the pheatmap package. Alignment files produced using IPD were used to compute the physical genome coverage of specific pathogens using SAMtools (30) . Total microbiome quantification, representing archaea, bacteria, plasmids, viruses, fungi and protozoa, was done using Kraken2 (31) . The read counts from Kraken2 were normalized by converting the read count to reads per million for each sample and are termed as total microbiome burden (TMiB). HNSC tumor samples (in-house and TCGA-HNSC) were sorted based on the HPV burden. Top and bottom 10% tumors were assigned status as HPV-high and -low, respectively. In the case of matching values at the high and low boundaries, all the tumors having matching values were assigned the same status. A similar assignment of high and low status was done using the F. nucleatum burden. Tumors for which both HPV and Fusobacterium status was assigned were used for the mutual exclusivity analysis. Statistical significance for the mutual exclusivity was computed using CoMEt package (32) . The RNA-seq tier 1 data (aligned BAM) for TCGA-HNSC (n = 546) and TCGA-COAD (n = 512) project were obtained from the National Cancer Institute Cancer Genome Commons Portal (GDC; http://portal.gdc.cancer. gov). The BAM files were converted to raw fastq files using the SamToFastq utility of the Picard toolkit (https: //broadinstitute.github.io/picard/). The raw fastq data were analyzed further for pathogen quantification using IPD. The htseq-count files for individual samples for RNAseq and miRNA expression data were downloaded from the GDC. Immune and leukocyte fraction data for individual samples were downloaded from the GDC (https:// gdc.cancer.gov/about-data/publications/panimmune). The neutrophil-to-lymphocyte ratio (NLR) was computed by dividing the neutrophil fraction by the aggregated lymphocyte fraction [as defined by CIBERSORT (33)] within individual samples. The differential gene and miRNA expression analysis was performed using the DESeq2 package (34) . The false discovery rate of <0.05 was considered significant, and genes with log 2 fold change ≥1.5 or ≤−1.5 were defined as significantly up-and downregulated, respectively. For miRNA comparisons, log 2 fold changes of ≥0.5 and ≤−0.5 were defined as significantly up-and downregulated, respectively. Pathogen abundance comparison between the cancer types, immune fraction and mutation burden comparison between a group of samples (subtypes of samples based on pathogen abundance) were performed using the Wilcoxon test (Mann-Whitney), where factors showing Pvalue <0.01 were considered significantly different. RSEM quantification of the MMP10 gene was extracted from the TCGA-HNSC matrix from the GDC. Comparison between the subgroups for the expression was performed using the Wilcoxon test. The survival analysis was performed using the KMPlot web server (35) . Fusobacterium status was assigned to the samples assessed in the survival analysis based on the 'Auto select' option, which selects the bestperforming cutoff between the upper and lower quartiles. The following primer sequences were used for Fusobacterium validation as reported earlier (36) . PCR was car-ried out in a 10 l reaction volume containing 5 l KAPA 2× ReadyMix Master Mix (Kapa Biosystems; cat. no. KK1024), 10 pmol of each primer and 20 ng of cDNA. The PCR conditions were as follows: an initial denaturation step at 94 • C for 2 min, 35 cycles of denaturation at 94 • C for 30 s, hybridization at 55 • C for 30 s, elongation at 72 • C for 30 s and a final extension step at 72 • C for 5 min on a PCR machine (HiMedia Prima-Duo™ Thermal Cycler, two blocks of 48 wells; LA948). Sanger sequencing was performed to confirm the presence of Fusobacterium in the samples. Total RNA was extracted from tongue cancer patient samples using AllPrep DNA/RNA/miRNA Universal Kit (QI-AGEN, cat. no. 80224) as per the manufacturer's protocol. Extracted RNA was resolved on 1.2% agarose gel to confirm the RNA integrity. DNase treatment was done using a DNA-free™ kit (Ambion, Foster City, CA, USA; cat. no. AM1906). For analyzing transcript levels of F. nucleatum and miRNA target genes, cDNA was synthesized with 500 ng of total RNA using high-capacity cDNA reverse transcription kit (Applied Biosystems, cat. no. 4368814). For miRNA quantification, cDNA synthesis was carried out with 500 ng of total RNA using pooled miRNA-specific stem-loop RT primers [protocol as described (37)]. Quantitative real-time PCR was performed on QuantStudio5 Real-Time PCR instrument (Thermo Fisher Scientific) using Mir-X miRNA qRT-PCR SYBR Master Mix (Clontech Takara, cat. no. 639676) for miRNAs and KAPA SYBR Fast Universal Mix (KAPA Biosystems, cat. no. KK4601) for Fusobacterium and miRNA target genes. Expression of candidate miRNAs and genes was calculated as CT. RNU48 was used as an internal control for microRNAs and beta-actin for Fusobacterium and genes. A set of 99 oral patient samples (tumor = 74, adjacent normal = 25) was used to perform real-time PCR-based quantification of F. nucleatum and validate the deregulated molecular factors associated with the pathogen levels. The validation set included 19 tumor-normal paired patient samples. qPCR-based levels of F. nucleatum were determined in the 74 tumors and the top and bottom quantiles (n = 18 in each group) of tumor samples were used as Fusobacterium-high and -low tumor samples, respectively. Gene and miRNA quantities were compared between these two groups of patient samples (n = 36 tumors) and significance was computed using the Wilcoxon rank-sum test. We reanalyzed TCGA and in-house generated 239 whole exome and 1168 transcriptome samples representing breast (38) , lung (27), colorectal (28), gallbladder (39), head and neck (40) , and cervical cancers (in-house sample statistics is provided in Supplementary Table S2) using IPD. IPD allows normalized quantification (scaled by FPM) for 1060 infectious pathogens, including the most common cancerassociated bacteria such as B. fragilis, H. pylori and F. nucleatum, and viruses such as HPV, Epstein-Barr virus, hepatitis C virus and others. HPV has been characterized as an etiological factor in cervical cancer (41) and is considered as an independent risk factor in oropharyngeal tumors (42) . However, studies from our lab and others have reported no association of HPV with the Indian oral tumors (40, (43) (44) . Our analysis of the exome samples reiterates that there is no evidence of the presence of HPV in the in-house oral cancer samples (n = 46); however, the prevalence of HPV 16 and related strains was found in 44.18% cervical tumors (range Figure S1 ). The abundance of these and other bacteria has been confirmed by metagenomic sequencing studies in head and neck tumors (19, 47) . From the transcriptome dataset of the breast, cervical, colorectal, and head and neck cancer samples analyzed using IPD, distinct pathogen signatures were observed among the tumors (Figure 1 ). Since the transcriptome dataset used in the analysis is generated using two different capture methodologies (polyA and ribo-depleted), we checked for the difference in the pathogen quantification due to sequencing method/protocol. The total microbial read count generated by Kraken2 was found to be comparable across the tumors sequenced using two capture methods (Supplementary Figure S2A and B). Further principal component analysis (PCA) was performed to assess any batch effect due to sequencing protocols in the data. However, no specific clustering of the samples was observed based on the IPD-based pathogen read counts (Supplementary Figure S3) . Among the tumor types analyzed, breast tumors were found to bear the least infectious pathogen burden for the 1060 pathogens quantified. Recent studies have reported enrichment of Fusobacterium in breast tumors (48) ; however, we did not find similar enrichment in our in-house breast transcriptome samples. We further computed the TMiB (as described in the 'Materials and Methods' section), which indicates the proportion of captured pathogen reads among the reads sequenced per sample. The in-house breast transcriptome samples showed comparable TMiB to other tumor types, suggesting that the low Fusobacterium burden certainly depicts the absence of pathogen and may not be due to lack of capture of overall microbiota in these tu-mors (Supplementary Figure S4A and B) . Among the transcriptome of cervical tumors, expression of HPV was observed in 90.48% samples (range = 0-325.79 FPM), consistent with earlier reports (49) . The common inflammatory bacterium observed in the DNA samples was also found to be expressed in the transcriptome data across the cervical, colorectal, and head and neck tumors. Colorectal tumors form a subgroup associated with the overabundance of Fusobacterium (12, 36) . In the HNSC (n = 512) tumor samples, which included in-house oral and TCGA-HNSC samples, we observed the high abundance of HPV (abundance >1 FPM) in 12% (n = 63) of the HNSC tumors, consistent with earlier reports (50) . HPV is one of the prominent etiological factors of oropharyngeal tumors (51) and is known as the cancercausing agent in head and neck tumors (52) . Among the known pathogens associated with HNSC, Fusobacterium traces were present in 75.9% of HNSC tumors (range = 0-98.66 FPM, mean = 2.35 FPM), comparable to the colorectal tumors (78.2%). We observed that HPV and Fusobacterium formed distinct clustering of samples in the HNSC tumor. By using the tumor samples having the pathogen high and low status defined (described in the 'Materials and Methods' section), mutual exclusivity was calculated for the two pathogens. Interestingly, HPV and Fusobacterium were found to be mutually exclusive in the HNSC tumors (P-value = 0.0146). A map depicting overlap and prevalence of the four major pathogens (HPV, Fusobacterium, Bacteroides and pathogenic E. coli IAI39), across the transcriptome dataset, has been summarized in Supplementary Figure S5A and D. We further evaluated whether the Fusobacterium-rich subgroup of tumors also formed a distinct molecular and/or clinical subtype of the HNSC tumors. As various HPV strains are known to impart a significant influence over the gene and immune expression profile in head and neck cancer (53) , all the tumors showing HPV expression >1 FPM were annotated as HPVpositive. To correlate Fusobacterium abundance with molecular and clinical features, we used the 438 HPV-negative tumor samples from TCGA-HNSC. These were then divided into Fusobacterium-high and -low, by sorting the samples based on pathogen burden gradient. Of these, 133 tumors showed no expression of F. nucleatum and were considered to be Fusobacterium-low and the top 10% quantile (n = 44) as Fusobacterium-high. We performed differential expression analysis between these two sub- Table S3 ). Reactome (54)based overrepresentation analysis of the upregulated genes showed keratinization/cornified envelope formation (Pvalue = 7.88e−15) and interleukin signaling (P-value = 7.88e−15) as the top enriched pathways, which are also reported to be primarily activated in the oral epithelium during bacterial dysbiosis and inflammation (55) (Supplementary Table S4 ). To get a comparative account of the pathogen-induced expression profile change, a similar analysis was performed between HPV-positive versus -negative TCGA-HNSC tumors. We observed 209 upregulated and 553 downregulated genes in HPV-positive tumors. Comparing the list of deregulated genes among Fusobacteriumbased and HPV-based differential analysis, 34 genes were found to be commonly downregulated ( Supplementary Figure S6A) ; however, no gene was commonly upregulated among the two subgroups (Supplementary Figure S6B ). This suggests a distinct role of the two pathogens in the modulation of the HNSC tumor gene expression profile. Among those differentially expressed, MMP10, a potential biomarker for prediction of nodal metastases in oral cancer (unpublished data), was found to be significantly higher in the Fusobacterium-high subgroup (Supplementary Figure S7 ), suggesting a potential association between the Fusobacterium load and nodal metastases. Additionally, multiple inflammatory pathway genes (n = 118) were found to be upregulated (adjusted P-value <0.05) in the Fusobacterium-high TCGA-HNSC subgroup. Among the inflammatory pathway genes, we selected IL1B, IL6 and IL8 as representative effector molecules for the real-time qPCR validation. The in-house validation sample set consisted of clinically assessed early-stage tongue tumors. In the validation cohort (n = 74), real-time qPCR of 16S RNA gene of Fusobacterium was performed and the upper and lower quantile tumors were stratified as Fusobacterium-high and -low, based on their pathogen load (described in the 'Materials and Methods' section). There were 19 tumornormal pairs in the validation sample set, in which we compared the levels of Fusobacterium. However, no difference was observed in its levels in the tumor and normal samples (Supplementary Figure S8) . Analysis showed that the three selected pro-inflammation markers (IL1B, IL8, IL6) were significantly higher in the Fusobacteriumhigh tumor samples (Figure 2A-C) , suggesting elevated levels of inflammation in these tumors. Being inflammatory response mediators, overexpression of TLR4 and NFκB is reported in response to Fusobacterium infection in colorectal tumors (56) . However, we observed no difference in the mRNA expression levels of NFκB and TRL4 in the in-house Fusobacterium-high and -low tongue tumor subgroups ( Figure 2D and E), unlike colorectal cancer. Inflammatory pathways are also mediated by the expression of specific miRNAs and have been considered as useful biomarkers in cancer, as well as pathogenic infections (57) . In the TCGA-HNSC HPV-negative tumors, we performed differential miRNA expression analysis between the Fusobacterium-high (n = 44) versus Fusobacterium-negative (n = 130) tumors. Similarly, differential miRNA expression analysis was also performed between the TCGA-HNSC tumor versus normal, and HPV-positive versus -negative tumor samples. In total, 62 miRNAs (30 upregulated and 32 downregulated) were identified to be deregulated (Supplementary Table S5 ) in the Fusobacterium-based comparison. Of the 62, by overlapping with the tumor-normal and HPV positive-negative differential miRNA lists, we identified 12 upregulated and 11 downregulated miRNAs that were specific to the Fusobacterium-high subgroup of the HNSC tumors ( Figure 3A and B) . We selected top five upregulated (hsa-mir-451a, hsa-mir-675, hsa-mir-203a, hsa-mir-144, hsamir-486-1) and three downregulated (hsa-mir-1269b, hsamir-9, hsa-mir-598) miRNAs for validation in the in-house Table 1 ). Of the upregulated miRNAs, expression levels of hsa-mir-451a, hsa-mir-675 and hsa-mir-486-1 were found to be significantly elevated in the Fusobacterium-high subgroup of tongue tumors ( Figure 3C -E), whereas others were invalidated. Individually, the three validated miRNAs have previously been reported to be involved in inflammation (58) (59) (60) . Additionally, the experimentally validated targets of these miRNAs have also been reported to be directly involved in the inflammatory pathway. For example, has-mir-451a targets major genes in the inflammatory pathway, such as MIF (61), AKT1 (62), MYC (63) and IKBKB (64), among others. CD40, a major mediator of inflammation response toward bacterial infection, is a known target of hsa-mir-486-1 (65) . However, hsa-mir-675 targets RUNX1 (66), a major suppressor of TLR4-based inflammatory response (67) . This suggests direct as well as indirect involvement of the three validated miRNAs in inflammatory response management. Overall, we identified several Fusobacteriumassociated miRNAs in the head and neck tumors and validated three hsa-mir-451a, hsa-mir-675 and hsa-mir-486-1 to be positively correlated with Fusobacterium burden. Fusobacterium abundance has been shown to influence the tumor immune microenvironment and gene expression profile in colorectal tumors (68, 69) . Gene expression-based approaches, such as CIBERSORT (33) , are used to decipher the fraction of individual immune cells from tumor environment. We thus obtained the precomputed, CIBERSORTbased immune cell fraction scores for the TCGA-HNSC tumor samples from the GDC and compared their levels between the Fusobacterium-high and -low TCGA-HNSC subgroups. Among the various immune cell types, activated mast cell (P-value = 6.2e−07), neutrophil (P-value = 0.018), activated dendritic cell (P-value = 0.031) and M2 macrophage (P-value = 0.035) fractions (scores) were enriched, whereas regulatory T-cell (P-value = 9.8e−04) fractions were significantly low in the Fusobacterium-high subgroup (Figure 4) . There was no significant difference observed between the major antitumor immune cell (CD8 + and CD4 + T cells and B cells) composition between the two subgroups, which is in contrast to colorectal tumors (70) . Based on the immune cell enrichment scores, we also computed the NLR, which is a marker for systemic in- flammation and an important prognostic marker in head and neck tumors (71) . NLR was significantly found to be higher in the Fusobacterium-high (P-value = 0.0084) TCGA-HNSC subgroup (Supplementary Figure S9) . Immune signature analysis indicates that Fusobacterium load is associated with an inflamed, innate immune cell-enriched and potentially pro-tumorigenic microenvironment, as opposed to the HPV-positive HNSC tumors that display T-cell inflamed phenotype (72) . Since the Fusobacterium levels are shown to have elevated inflammation and a distinct immune profile, we further evaluated its possible relation with clinical features and patient survival. From the validation cohort (detailed in Table 1) of the in-house tongue tumors, the clinical feature and survival details were available for 52 of the 74 patients screened for Fusobacterium. For these patients, the Fusobacterium-high and -low status was assigned based on the best-performing CT value cutoff of KMPlot. Com-paring the overall survival, the in-house patient samples showed that Fusobacterium-high patients showed poor survival (log-rank P-value = 0.016) ( Figure 5A ). Since all the in-house patient samples were of tongue origin, we segregated the TCGA-HNSC HPV-negative tumors based on the defined anatomical site and performed survival analysis using the patients with tumor of tongue origin (n = 107). Consistent with the observation from the in-house tumor samples, the TCGA tongue cancer patients with Fusobacteriumhigh status showed a trend of poor overall survival (hazard ratio = 1.67, log-rank P-value = 0.14) ( Figure 5B ). We further applied Cox proportional hazard model to describe risk factors. In addition to Fusobacterium load status, alcohol usage, smoking status, tobacco usage, nodal status, disease recurrence status, perineural invasion, lymphovascular invasion and extracapsular spread status were used as covariates. Among these, Fusobacterium-high status (2.66e+01), smoking (5.24), tobacco (1.34e+01), extracapsular spread (2.59e+01) and disease recurrence status (4.02e+01) showed positive regression coefficients, although none of the factors was statistically significant. Next, we performed association analysis between the Fusobacterium burden status (as assigned by KMPlot) with lymph node positivity, lymphovascular invasion, perineural invasion, extracapsular spread and recurrence status of the in-house tongue tumors. Among these, higher Fusobacterium was associated with the presence of extracapsular spread (Fisher test; odds ratio = 4.98, P-value <0.05). Fusobacterium load was not found to be associated with alcohol consumption, tobacco smoking or chewing status in the patient cohort. Overall, Fusobacterium burden was found to be associated with poor prognosis and higher extracapsular spread, especially in tongue tumor cancer. Pathogen-induced inflammation is a major factor determining the disease outcome and response to therapy (73, 74) . Several pathogens have been categorized as cancer risk factors in different cancers, with studies emphasizing the role of periodontal pathogens in head and neck cancer. Enrichment of specific bacterial populations has been associated with tumors, mainly using metagenomic sequencing datasets. Recently, identification of viral and bacterial pathogens has also been performed from the human DNA and RNA sequencing data, using varied computational tools (22) . In this study, we systematically analyzed 239 whole exome and 1168 whole transcriptome sequencing datasets representing six tumor types: breast, cervical, colorectal, lung, gallbladder, and head and neck cancers, to quantify the abundance of 1060 pathogens using our previously published method (IPD). This method allows us to generate a normalized quantification for diverse cancer-associated pathogens among different tumor types, directly from the NGS datasets. From the landscape of pathogens identified across the sequencing dataset, we observed traces of known cancer-associated pathogens such as HPV in cervical and HNSC tumors, B. fragilis and pathogenic E. coli in colorectal tumors, and F. nucleatum in colorectal and HNSC tumors. Interestingly, Fusobacterium persistence was found to be in the comparable frequency in colorectal and HNSC tumors. In the HNSC transcriptome samples, HPV was found to be forming a distinct subtype. HPV-associated HNSC tumors have been previously characterized and are reported to form a distinct clinical and molecular subtype (50) . Specifically, in the HPV-negative oral tumors, periodontal pathogens have been reported to be a risk factor (21) . In our analysis, we observed that Fusobacterium-high tumors formed another distinct subgroup in HNSC, which was mutually exclusive to the HPVhigh subgroup. Fusobacterium has been primarily reported to be associated with poor prognosis and recurrence in colorectal tumors (36, 75) . Along with other inflammationinducing pathogens, it has also been reported to be significantly enriched in head and neck tumors (19, 76) . Further, only considering the HPV-negative TCGA-HNSC samples (n = 438), we classified tumors having top 10% pathogen load as Fuso-high and with no expression as Fuso-negative. To unveil the molecular and clinical features associated with Fusobacterium burden in HNSC, we performed differential gene expression analysis between the Fusobacteriumhigh versus -low tumors. The significantly upregulated genes were strikingly different from the genes upregulated in HPV-positive HNSC tumors. Fusobacterium-high tumors had 118 inflammatory pathway genes upregulated, suggesting an inflamed tumor microenvironment. Among these, MMP10, a biomarker for lymph node metastases in oral tumors (unpublished data), was also found to be positively associated with Fusobacterium burden in TCGA-HNSC tumors. This suggests a possible role of Fusobacterium in nodal metastases in oral tongue tumors by induction of chronic inflammation. To further validate the association between Fusobacterium burden and inflammation, we evaluated the levels of pro-inflammatory cytokines IL1B, IL8 and IL6 in Fusobacterium-high and -low inhouse early-stage tongue tumor samples (n = 75). Expression of these cytokines was found to be significantly higher in the tongue tumors having a higher load of Fusobacterium. These inflammation markers are known to get activated upon TLR signaling. Monolayer co-infection experiments have shown that Fusobacterium induction enhances TLR signaling in cell line and mouse models (77, 78) . However, the transcript levels of NFκB and TLR4 remain unaltered in Fusobacterium-high tongue tumors, unlike colorectal tumors wherein the activity as well as transcriptional elevation is observed upon Fusobacterium infection. Inflammation is also known to be tightly regulated by the miRNA expression profile in the backdrop of infection and cancer. We identified differentially expressed miR-NAs in Fusobacterium-high versus -negative, HPV-negative TCGA-HNSC tumors and compared the list of deregulated miRNAs with the ones associated with HPV and HNSC tumor only. From the miRNAs deregulated specifically in the Fusobacterium-high group, we selected eight (five upregulated and three downregulated) miRNAs for validation in the in-house tongue cancer patient cohort. Of these, three upregulated miRNAs miR-451a, miR-675 and miR-486-1 were validated to have significantly higher expression in the Fusobacterium-high tongue tumors. These three miRNAs have been previously independently reported to be biomarkers of progression, treatment response and disease prognosis in different tumor types and have also been linked to inflammation (58) (59) (60) 79) . Viral infection and bacterial dysbiosis have been linked to a distinct immune profile of the tumors. Very little is known about the HPV-negative HNSC tumors in terms of the immune cell composition and function (80) . We evaluated the immune cell composition of the HPV-negative HNSC tumors and observed enrichment of innate immunity cell types, including neutrophils, M2 macrophages, mast cells and dendritic cells, in tumors with a higher burden of Fusobacterium. These tumors were also found to have a higher NLR, indicating an inflamed state. The expression analysis of the inflammation-related genes and immune cell enrichment analysis suggest Fusobacterium burden is a highly inflamed, pro-tumorigenic tumor microenvironment. Further, we performed a correlation of the Fusobacterium burden with various clinical features. We primarily observed that a higher burden of the pathogen is associated with the lower overall survival in in-house tongue cancer patients. We extended the analysis to the HPV-negative TCGA oral tongue tumor patients and observed a marginally significant difference in the survival, consistent with the inhouse tongue cancer samples. We also performed an association analysis of the pathogen burden with different clinical features, including perineural invasion, lymph node pre-sentation, and extracapsular and lymphovascular spread. Among these, we observed a significant association of Fusobacterium burden with the presence of extracapsular spread in tongue cancer patients. This suggests that a higher Fusobacterium burden is a poor prognosis factor and may be associated with higher invasion and metastasis, especially in patients with tongue tumors. Since the clinical association and validation patient samples were primarily clinically assessed for early-stage tumors, this study highlights role of Fusobacterium in the early-stage tongue tumors, which are on the rise in the developing countries. The role of Fusobacterium in tongue cancer may follow the disease progression models implicated in colorectal and pancreatic cancers, with tumor type and site-specific presentation of the molecular features. Recent studies have also shown Fusobacterium burden to be associated with recurrence, smoking and tobacco chewing status among other clinical features in head and neck cancer (81) . In our samples, we did not observe these associations, probably because of the smaller cohort size. Another considerable deficiency of this study is unavailability of clinical information regarding treatment history and pre-operative prophylactic antibiotic treatment, if any. Since both of these factors may influence the prevalence of microbes in the tumor microenvironment, inclusion of this information may further add significance, also with respect to the disease outcome in the context of prevailing infection. Additionally, capture of pathogens from the human exome sequencing is primarily dependent on the nonspecific crosshybridization of the pathogen DNA with the probes. Also, the different types of RNA sequencing capture kits (such as polyA and ribo-depletion) may have differential sensitivities for detecting the microbial transcripts, affecting their quantification in the computational subtraction. Finally, the Fusobacterium burden and its tolerance across ethnicities may be driven by oral hygiene, immunity, geographical and dietary factors, which may present a different outcome in different populations. Our study presents a comprehensive landscape of pathogens identified from the genomic dataset of Indian origin, across six different tumor types. Combining the TCGA data, we identify a subgroup of poor prognosis tongue tumors, which may be primarily driven by the inflammation due to pathogens such as Fusobacterium and points to important clinical implications of the burden of the periodontal pathogens, especially in HPV-negative head and neck cancer patients. This may provide a rationale to further design studies to understand the variability in treatment response, disease recurrence and resistance, and adoption of additional measures such as the use of antibiotics for tumors of specific origin, similar to colorectal cancer. All the genomic datasets analyzed in the manuscript can be accessed from the ArrayExpress database under the accession numbers E-MTAB-11412, E-MTAB-9766, E-MTAB-6619, E-MTAB-4653, E-MTAB-8801, E-MTAB-11404, E-MTAB-9281 and E-MTAB-11407. The colorectal cancer data have been deposited at the European Genome-phenome Archive, under accession number EGAS00001005970. Microbe-microbe and host-microbe interactions drive microbiome dysbiosis and inflammatory processes The oral microbiota: living with a permanent guest Cancer and the microbiota Mutational signature in colorectal cancer caused by genotoxic pks + E. coli Microbes, microbiota, and colon cancer Lejondal,H. and pylori-Gastric Cancer Task Force (2005) Helicobacter pylori eradication has the potential to prevent gastric cancer: a state-of-the-art critique Helicobacter pylori infection induces gastric cancer in Mongolian gerbils Helicobacter pylori but not high salt induces gastric intraepithelial neoplasia in B6129 mice Bacteria and cancer: cause, coincidence or cure? A review Non-typhoidal Salmonella DNA traces in gallbladder cancer Promotion of intestinal carcinogenesis by Streptococcus bovis Genomic analysis identifies association of Fusobacterium with colorectal carcinoma A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorectal cancer Association between intraepithelial Escherichia coli and colorectal cancer Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma Chlamydia pneumoniae and lung cancer: epidemiologic evidence The oralome and its dysbiosis: new insights into oral microbiome-host interactions Oral microbiota community dynamics associated with oral squamous cell carcinoma staging The oral microbiota may have influence on oral cancer 2020) Detection of Fusobacterium in oral and head and neck cancer samples: a systematic review and meta-analysis Periodontal pathogens are a risk factor of oral cavity squamous cell carcinoma, independent of tobacco and alcohol and human papillomavirus Tissue-associated microbial detection in cancer using human sequencing data Adding pathogens by genomic subtraction 2021) An integrated approach to determine the abundance, mutation rate and phylogeny of the SARS-CoV-2 genome The microflora associated with human oral carcinomas Extent of extracapsular spread: a critical prognosticator in oral tongue cancer Molecular characterization of lung squamous cell carcinoma tumors reveals therapeutically relevant alterations Exome sequencing identifies ARID2 as a novel tumor suppressor in early-onset sporadic rectal cancer The landscape of viral associations in human cancers The Sequence Alignment/Map format and SAMtools Improved metagenomic analysis with Kraken 2 CoMEt: a statistical approach to identify combinations of mutually exclusive alterations in cancer Robust enumeration of cell subsets from tissue expression profiles Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 Web-based survival analysis tool tailored for medical research (KMplot): development and implementation Analysis of Fusobacterium persistence and antibiotic response in colorectal cancer Stem-loop RT-qPCR for miRNAs Pre-operative progesterone benefits operable breast cancer patients by modulating surgical stress ERBB2 and KRAS alterations mediate response to EGFR inhibitors in early stage gallbladder cancer Genomic characterization of tobacco/nut chewing HPV-negative early stage tongue tumors identify MMP10 as a candidate to predict metastases The causal link between human papillomavirus and invasive cervical cancer: a population-based case-control study in Colombia and Spain Human papillomavirus as a driver of head and neck cancers NGS-based approach to determine the presence of HPV and their sites of integration in human cancer genome No role for human papillomavirus infection in oral cancers in a region in southern India The Bacteroides fragilis toxin gene is prevalent in the colon mucosa of colorectal cancer patients Bacterial infection profiles in lung cancer patients with febrile neutropenia Inflammatory bacteriome featuring Fusobacterium nucleatum and Pseudomonas aeruginosa identified in association with oral squamous cell carcinoma Breast cancer colonization by Fusobacterium nucleatum accelerates tumor growth and metastatic progression HPV genotypes distribution in Indian women with and without cervical carcinoma: implication for HPV vaccination program in Odisha, Eastern India The Cancer Genome Atlas Network (2015) Comprehensive genomic characterization of head and neck squamous cell carcinomas Case-control study of human papillomavirus and oropharyngeal cancer Evidence for a causal association between human papillomavirus and a subset of head and neck cancers Tumor microenvironment characterization in head and neck cancer identifies prognostic and immunotherapeutically relevant gene signatures The Reactome pathway knowledgebase The maintenance of an oral epithelial barrier Fusobacterium nucleatum increases proliferation of colorectal cancer cells and tumor development in mice by activating Toll-like receptor 4 signaling to nuclear factor-kappaB, and up-regulating expression of microRNA-21 MicroRNA as biomarkers and diagnostics 2017) miR-675 promotes disease progression of non-small cell lung cancer via activating NF-kappaB signaling pathway Circulating microRNAs as potential cancer biomarkers: the advantage and disadvantage Identification of microRNA-451a as a novel circulating biomarker for colorectal cancer diagnosis MicroRNA-451 regulates macrophage migration inhibitory factor production and proliferation of gastrointestinal cancer cells ) miRNA-451 plays a role as tumor suppressor in human glioma cells Repression of tumor suppressor miR-451 is essential for NOTCH1-induced oncogenesis in T-ALL ) miR-451 inhibits cell proliferation in human hepatocellular carcinoma through direct suppression of IKK-beta CD40 ligand mediates inflammation independently of CD40 by interaction with Mac-1 The long non-coding RNA H19-derived miR-675 modulates human gastric cancer cell proliferation by targeting tumor suppressor RUNX1 Runx1 negatively regulates inflammatory cytokine production by neutrophils in response to Toll-like receptor signaling Fusobacterium nucleatum and T cells in colorectal carcinoma Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment Fusobacterium nucleatum in colorectal cancer relates to immune response differentially by tumor microsatellite instability status Neutrophil-to-lymphocyte ratio in head and neck cancer prognosis: a systematic review and meta-analysis Treatment-naïve HPV+ head and neck cancers display a T-cell-inflamed phenotype distinct from their HPV− counterparts that has implications for immunotherapy Inflammation and tumor progression: signaling pathways and targeted intervention Fusobacterium nucleatum persistence and risk of recurrence after preoperative treatment in locally advanced rectal cancer Variations in oral microbiota associated with oral cancer Periodontal pathogens Porphyromonas gingivalis and Fusobacterium nucleatum promote tumor progression in an oral-specific chemical carcinogenesis model Fusobacteria modulate oral carcinogenesis and promote cancer progression Comprehensive upstream and downstream regulatory analyses identify miR-675-3p as a potential prognostic biomarker in melanoma Immunological network in head and neck squamous cell carcinoma--a prognostic tool beyond HPV status Prognostic value of intratumoral Fusobacterium nucleatum and association with immune-related gene expression in oral squamous cell carcinoma patients We thank all members of the Dutt laboratory for critically reading the manuscript. S.D. and A.J. Supplementary Data are available at NAR Cancer Online.