Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Amending microbiota by targeting intestinal inflammation with TNF blockade attenuates development of colorectal cancer

Abstract

Intestinal inflammation and microbiota are two important components of colorectal cancer (CRC) etiology. However, it is not clear how tuning inflammation using clinically relevant anti-inflammatory treatment impacts microbiota or whether this consequently influences CRC outcome. Here, using chemically induced (DSS/Apcmin/+) and spontaneous (Apcmin/+;Il10−/−) mouse CRC models colonized by colibactin-producing Escherichia coli, we established the role of microbiota in mediating the antitumorigenic effect of anti–tumor necrosis factor (TNF) therapy. We found that TNF blockade attenuated colitis and CRC development. Microbiota community structure and gene activities significantly changed with disease development, which was prevented by TNF blockade. Several microbiota functional pathways underwent similar changes in patients following anti-TNF therapy. Under cohousing condition, TNF blockade failed to prevent colitis, cancer development and disease-associated microbiota structural changes. Finally, microbiota transplantation showed reduced carcinogenic activity of microbiota from anti-TNF-treated mice. Together, our data demonstrate the plasticity of microbiota, which could be reverted to noncarcinogenic status by targeting inflammation.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: E. coli NC101 promotes CRC development via colibactin activity in mice harboring a complex microbiota.
Fig. 2: TNF blockade attenuates NC101-mediated CRC development.
Fig. 3: TNF blockade alters microbiota composition in DSS/Apcmin/+ mice.
Fig. 4: TNF blockade alters microbiota activity in DSS/Apcmin/+ mice.
Fig. 5: Cohousing abrogates TNF blockade effect on microbiota, inflammation and CRC.
Fig. 6: TNF blockade-associated microbiota show reduced carcinogenic activity.

Similar content being viewed by others

Data availability

The sequencing data generated in this study were deposited in the National Center for Biotechnology Information Sequence Read Archive under the following accessions: PRJNA564272 (transplant 16S rDNA sequences), PRJNA564144 (cohousing 16S rDNA sequences), PRJNA564137 (main experiment 16S rDNA sequences), PRJNA564115 (microbial RNA-seq) and PRJNA610017 (metagenome shotgun sequences). The human metatranscriptome and metagenome data were derived from the IBDMDB (https://ibdmdb.org/), and the unique ‘External ID’ for individual samples included in our analysis was listed in Supplementary Table 5. Data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

Code availability

The Leica Application Suite X (https://www.leica-microsystems.com/products/microscope-software/p/leica-las-x-ls/) was used for histology and immunohistochemistry visualization and imaging. The following open-source code was used in the analyses performed in this research: QIIME v.1.9.1 (ref. 51), Trimmomatic v.0.36 (ref. 57), KneadData v.0.6.1 (http://huttenhower.sph.harvard.edu/kneaddata), Bowtie2 v.2.3.5 (ref. 58), featureCounts v.1.5.3 (ref. 59), HUMAnN2 v.2.8.1 (ref. 60), edgeR v.3.26, GAGE v.2.34 (ref. 61), Diamond v.0.9.24 (ref. 63), R statistical framework v.3.5.1, R phyloseq package v.1.26, R nlme package v.3.1-140 and R ggplot2 package v.3.2. In addition, the following commercial code was used for data analyses: COSMOSID (https://www.cosmosid.com/) and GraphPad Prism 8 (https://www.graphpad.com/scientific-software/prism/). Preprocessing steps were done using in-house scripts that are available upon request.

References

  1. Ng, S. C. et al. Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet 390, 2769–2778 (2018).

    Google Scholar 

  2. Beaugerie, L. & Itzkowitz, S. H. Cancers complicating inflammatory bowel disease. N. Engl. J. Med. 372, 1441–1452 (2015).

    CAS  PubMed  Google Scholar 

  3. Greten, F. R. & Grivennikov, S. I. Inflammation and cancer: triggers, mechanisms, and consequences. Immunity 51, 27–41 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Schirmer, M., Garner, A., Vlamakis, H. & Xavier, R. J. Microbial genes and pathways in inflammatory bowel disease. Nat. Rev. Microbiol. 17, 497–511 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Kang, M. & Martin, A. Microbiome and colorectal cancer: unraveling host-microbiota interactions in colitis-associated colorectal cancer development. Semin. Immunol. 32, 3–13 (2017).

    PubMed  Google Scholar 

  6. Nougayrède, J.-P. et al. Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science 313, 848–851 (2006).

    PubMed  Google Scholar 

  7. Wilson, M. R. et al. The human gut bacterial genotoxin colibactin alkylates DNA. Science 363, eaar7785 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Xue, M. et al. Structure elucidation of colibactin and its DNA cross-links. Science 365, eaax2685 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Arthur, J. C. et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 338, 120–123 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Buc, E. et al. High prevalence of mucosa-associated E. coli producing cyclomodulin and genotoxin in colon cancer. PLoS ONE 8, e56964 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Prorok-Hamon, M. et al. Colonic mucosa-associated diffusely adherent afaC+ Escherichia coli expressing lpfA and pks are increased in inflammatory bowel disease and colon cancer. Gut 63, 761–770 (2014).

    CAS  PubMed  Google Scholar 

  12. Dejea, C. M. et al. Patients with familial adenomatous polyposis harbor colonic biofilms containing tumorigenic bacteria. Science 359, 592–597 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Eklöf, V. et al. Cancer-associated fecal microbial markers in colorectal cancer detection. Int. J. Cancer 141, 2528–2536 (2017).

    PubMed  PubMed Central  Google Scholar 

  14. Wirbel, J. et al. Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat. Med. 25, 679–689 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Pleguezuelos-Manzano, C. et al. Mutational signature in colorectal cancer caused by genotoxic pks + E. coli. Nature 580, 269–273 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Bonnet, M. et al. Colonization of the human gut by E. coli and colorectal cancer risk. Clin. Cancer Res. 20, 859–867 (2014).

    PubMed  Google Scholar 

  17. Cougnoux, A. et al. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut 63, 1932–1942 (2014).

    CAS  PubMed  Google Scholar 

  18. Arthur, J. C. et al. Microbial genomic analysis reveals the essential role of inflammation in bacteria-induced colorectal cancer. Nat. Commun. 5, 4724 (2014).

    CAS  PubMed  Google Scholar 

  19. Tomkovich, S. et al. Locoregional effects of microbiota in a preclinical model of colon carcinogenesis. Cancer Res. 77, 2620–2632 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Zhu, W. et al. Editing of the gut microbiota reduces carcinogenesis in mouse models of colitis-associated colorectal cancer. J. Exp. Med. 216, 2378–2393 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Brotherton, C. A. & Balskus, E. P. A prodrug resistance mechanism is involved in colibactin biosynthesis and cytotoxicity. J. Am. Chem. Soc. 135, 3359–3362 (2013).

    CAS  PubMed  Google Scholar 

  22. Cougnoux, A. et al. Small-molecule inhibitors prevent the genotoxic and protumoural effects induced by colibactin-producing bacteria. Gut 65, 278–285 (2016).

    CAS  PubMed  Google Scholar 

  23. Gogokhia, L. et al. Expansion of bacteriophages is linked to aggravated intestinal inflammation and colitis. Cell Host Microbe 25, 285–299.e8 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Winter, S. E. et al. Host-derived nitrate boosts growth of E. coli in the inflamed gut. Science 339, 708–711 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Hughes, E. R. et al. Microbial respiration and formate oxidation as metabolic signatures of inflammation-associated dysbiosis. Cell Host Microbe 21, 208–219 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Zhu, W. et al. Precision editing of the gut microbiota ameliorates colitis. Nature 553, 208–211 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Aden, K. et al. Metabolic functions of gut microbes associate with efficacy of tumor necrosis factor antagonists in patients with inflammatory bowel diseases. Gastroenterology 157, 1279–1292.e11 (2019).

    CAS  PubMed  Google Scholar 

  28. Martin, P. et al. Interplay between siderophores and colibactin genotoxin biosynthetic pathways in Escherichia coli. PLoS Pathog. 9, e1003437 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Karki, R. & Kanneganti, T.-D. Diverging inflammasome signals in tumorigenesis and potential targeting. Nat. Rev. Cancer 19, 197–214 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Estevinho, M. M. et al. Features of fecal and colon microbiomes associate with responses to biologic therapies for inflammatory bowel diseases: a systematic review. Clin. Gastroenterol. Hepatol. 18, 1054–1069 (2019).

    PubMed  Google Scholar 

  31. Lloyd-Price, J. et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature 569, 655–662 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Xiao, L. et al. A catalog of the mouse gut metagenome. Nat. Biotechnol. 33, 1103–1108 (2015).

    CAS  PubMed  Google Scholar 

  33. Zhang, Z., Chen, F. & Shang, L. Advances in antitumor effects of NSAIDs. Cancer Manag. Res. 10, 4631–4640 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Biancone, L. et al. Long-term safety of infliximab for the treatment of inflammatory bowel disease: does blocking TNFɑ reduce colitis-associated colorectal carcinogenesis? Gut 58, 1703 (2009).

    CAS  PubMed  Google Scholar 

  35. Baars, J. E. et al. The risk of inflammatory bowel disease-related colorectal carcinoma is limited: results from a nationwide nested case–control study. Am. J. Gastroenterol. 106, 319–328 (2011).

    PubMed  Google Scholar 

  36. Nyboe Andersen, N. et al. Association between tumor necrosis factor-α antagonists and risk of cancer in patients with inflammatory bowel disease. JAMA 311, 2406–2413 (2014).

    PubMed  Google Scholar 

  37. Rao, V. P. et al. Proinflammatory CD4+CD45RBhi lymphocytes promote mammary and intestinal carcinogenesis in Apc Min/+ mice. Cancer Res. 66, 57–61 (2006).

    CAS  PubMed  Google Scholar 

  38. Popivanova, B. K. et al. Blocking TNF-ɑ in mice reduces colorectal carcinogenesis associated with chronic colitis. J. Clin. Invest. 118, 560–570 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Onizawa, M. et al. Signaling pathway via TNF-ɑ/NF-κB in intestinal epithelial cells may be directly involved in colitis-associated carcinogenesis. Am. J. Physiol. Gastrointest. Liver Physiol. 296, G850–G859 (2009).

    CAS  PubMed  Google Scholar 

  40. Kim, Y. J., Hong, K. S., Chung, J. W., Kim, J. H. & Hahm, K. B. Prevention of colitis-associated carcinogenesis with infliximab. Cancer Prev. Res. (Phila.) 3, 1314–1333 (2010).

    CAS  Google Scholar 

  41. Craven, B., Zaric, V., Martin, A., Mureau, C. & Egan, L. J. Effect of genetic deletion or pharmacological antagonism of tumor necrosis factor alpha on colitis-associated carcinogenesis in mice. Inflamm. Bowel Dis. 21, 485–495 (2015).

    PubMed  Google Scholar 

  42. Lopetuso, L. R. et al. Infliximab does not increase colonic cancer risk associated to murine chronic colitis. World J. Gastroenterol. 22, 9727–9733 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Roda, G., Jharap, B., Neeraj, N. & Colombel, J.-F. Loss of response to anti-TNFs: definition, epidemiology, and management. Clin. Transl. Gastroenterol. 7, e135 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Bjarnason, I. et al. Mechanisms of damage to the gastrointestinal tract from nonsteroidal anti-inflammatory drugs. Gastroenterology 154, 500–514 (2018).

    CAS  PubMed  Google Scholar 

  45. Thomas, A. M. et al. Metagenomic analysis of colorectal cancer datasets identifies cross-cohort microbial diagnostic signatures and a link with choline degradation. Nat. Med. 25, 667–678 (2019).

    CAS  PubMed  Google Scholar 

  46. Chen, W., Liu, F., Ling, Z., Tong, X. & Xiang, C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS ONE 7, e39743 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Marchesi, J. R. et al. Towards the human colorectal cancer microbiome. PLoS ONE 6, e20447 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Yachida, S. et al. Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat. Med. 25, 968–976 (2019).

    CAS  PubMed  Google Scholar 

  49. Heaton, W. L. et al. Autocrine Tnf signaling favors malignant cells in myelofibrosis in a Tnfr2-dependent fashion. Leukemia 32, 2399–2411 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. He, Z. et al. Campylobacter jejuni promotes colorectal tumorigenesis through the action of cytolethal distending toxin. Gut 68, 289–300 (2019).

    CAS  PubMed  Google Scholar 

  51. Caporaso, J. G. et al. QIIME allows analysis of high-throughput community sequencing data. Nat. Methods 7, 335–336 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Bokulich, N. A. et al. Quality-filtering vastly improves diversity estimates from Illumina amplicon sequencing. Nat. Methods 10, 57–59 (2013).

    CAS  PubMed  Google Scholar 

  53. Wang, Q., Garrity, G. M., Tiedje, J. M. & Cole, J. R. Naive Bayesian classifier for rapid assignment of rRNA sequences into the new bacterial taxonomy. Appl. Environ. Microbiol. 73, 5261–5267 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. McMurdie, P. J. & Holmes, S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE 8, e61217 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. McCafferty, J. et al. Stochastic changes over time and not founder effects drive cage effects in microbial community assembly in a mouse model. ISME J. 7, 2116–2125 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Series B Stat. Methodol. 57, 289–300 (1995).

    Google Scholar 

  57. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    CAS  PubMed  Google Scholar 

  60. Franzosa, E. A. et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat. Methods 15, 962–968 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Luo, W., Friedman, M. S., Shedden, K., Hankenson, K. D. & Woolf, P. J. GAGE: generally applicable gene set enrichment for pathway analysis. BMC Bioinf. 10, 161 (2009).

    Google Scholar 

  62. Integrative HMP (iHMP) Research Network Consortium. The integrative human microbiome project. Nature 569, 641–648 (2019).

    Google Scholar 

  63. Buchfink, B., Xie, C. & Huson, D. H. Fast and sensitive protein alignment using DIAMOND. Nat. Methods 12, 59–60 (2015).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This research was supported by NIH grant R01DK073338, the University of Florida Health Cancer Center Funds and the University of Florida Department of Medicine Gatorade Fund (all to C.J.). Y.Y. was supported by the Crohn’s & Colitis Foundation of America research fellowship award (ref. no. 409472). R.Z.G. was supported by UF Health Cancer Center Funds. R.C.N. was supported by an NIH TL1 training grant (TL1TR001428). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript. We are grateful to the Germ-Free Services division of UF Animal Care Services for assistance with gnotobiotic experiments, the UF Molecular Pathology Core for assistance with tissue processing and staining, the UF Interdisciplinary Center for Biotechnology Research Core for assistance with shotgun metagenome and microbial RNA sequencing and J. Gauthier for assistance with 16S rRNA gene sequencing.

Author information

Authors and Affiliations

Authors

Contributions

Y.Y. and C.J. conceived and designed the study. Y.Y. and R.C.N. acquired data. Y.Y. and R.Z.G. analyzed data. Y.Y., R.Z.G. and C.J. drafted the manuscript. C.J. supervised the study. All authors reviewed and approved the manuscript.

Corresponding author

Correspondence to Christian Jobin.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 TNF blockade attenuates colonic inflammation and DNA damage response.

a, Timeline for short-term TNF blockade in NC101-colonized DSS/Apcmin/+ mice. Experiment was done once. b, Representative pictures of H&E stained colon Swiss roll section (distal). Scale bars = 500 μm. c, Distal colon histological scores. d, Representative pictures of colon Swiss roll sections stained for γH2AX. Scale bars = 100 μm. e, Association between treatment and incidence of γH2AX+ regenerating crypts. Fisher’s exact test was used to determine association. f, Fecal NC101 relative abundance in PBS- and anti-TNF-treated mice at day 14. g, Cytokine expression in distal colon snips of NC101-colonized Apcmin/+;Il10-/- mice treated with PBS or anti-TNF antibody (related to Fig. 2h). Data are mean ± s.d. Two-tailed Mann-Whitney U test was performed for paired comparisons. The number of animals (n) used for each graph was indicated.

Source data

Extended Data Fig. 2 Fecal microbiota and NC101 changes in DSS/Apcmin/+ mice treated with PBS or anti-TNF antibody.

a, Alpha diversity (Chao1) and beta diversity (PCoA) comparison of microbiota in PBS-treated mice between indicated time points. The box horizontal lines are the 25th (lower) and 75th (upper) percentile. The middle line is the 50th percentile (the median). The lower whisker is 25th percentile - 1.5 times the interquartile range and the upper whisker is 75th percentile + 1.5 times the interquartile range. b, Alpha diversity (Chao1) and beta diversity (PCoA) comparison of microbiota in anti-TNF-treated mice between indicated time points. Sequencing data analysis was described in details in Methods. The box horizontal lines are the 25th (lower) and 75th (upper) percentile. The middle line is the 50th percentile (the median). The lower whisker is 25th percentile - 1.5 times the interquartile range and the upper whisker is 75th percentile + 1.5 times the interquartile range. c, NC101 abundance by metagenome analysis. One-way ANOVA followed by Holm-Sidak’s multiple comparisons test was performed for multi-group comparisons. d, Normalized counts of transcripts aligned to clb genes. Two-tailed Mann-Whitney U test was performed for paired comparisons. Data are mean ± s.d. The number of animals (n) used for each graph was indicated.

Source data

Extended Data Fig. 3 Microbiota community structure changes in NC101-colonized Apcmin/+;Il10-/- mice treated with PBS or anti-TNF antibody.

a, Alpha diversity (Chao1) and beta diversity (PCoA) comparison of microbiota in PBS-treated mice between indicated time points. The box horizontal lines are the 25th (lower) and 75th (upper) percentile. The middle line is the 50th percentile (the median). The lower whisker is 25th percentile - 1.5 times the interquartile range and the upper whisker is 75th percentile + 1.5 times the interquartile range. b, Alpha diversity (Chao1) and beta diversity (PCoA) comparison of microbiota in anti-TNF-treated mice between indicated time points. The box horizontal lines are the 25th (lower) and 75th (upper) percentile. The middle line is the 50th percentile (the median). The lower whisker is 25th percentile - 1.5 times the interquartile range and the upper whisker is 75th percentile + 1.5 times the interquartile range. c, Alpha diversity (Chao1) and beta diversity (PCoA) comparison between PBS- and anti-TNF-treated mice in longitudinal stool samples and endpoint colon tissues. The box horizontal lines are the 25th (lower) and 75th (upper) percentile. The middle line is the 50th percentile (the median). The lower whisker is 25th percentile - 1.5 times the interquartile range and the upper whisker is 75th percentile + 1.5 times the interquartile range. ns, not significant. Sequencing data analysis was described in details in Methods. The number of animals (n) used for each graph was indicated.

Source data

Extended Data Fig. 4 Cohousing abolishes CRC-preventive effect of TNF blockade in NC101-colonized Apcmin/+;Il10-/- mice.

a, Schematic diagram for TNF blockade under cohousing condition. b, Macroscopic colon tumor counts. c, Distal colon histological scores. d, Fecal NC101 relative abundance in cohoused PBS- and anti-TNF-treated mice. Data are mean ± s.d. Two-tailed Mann-Whitney U test was performed for paired comparisons. Experiment was done once. The number of animals (n) used for each graph was indicated.

Source data

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, Y., Gharaibeh, R.Z., Newsome, R.C. et al. Amending microbiota by targeting intestinal inflammation with TNF blockade attenuates development of colorectal cancer. Nat Cancer 1, 723–734 (2020). https://doi.org/10.1038/s43018-020-0078-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-020-0078-7

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer