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.

Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B

Abstract

Acquired resistance to anticancer treatments is a substantial barrier to reducing the morbidity and mortality that is attributable to malignant tumors. Components of tissue microenvironments are recognized to profoundly influence cellular phenotypes, including susceptibilities to toxic insults. Using a genome-wide analysis of transcriptional responses to genotoxic stress induced by cancer therapeutics, we identified a spectrum of secreted proteins derived from the tumor microenvironment that includes the Wnt family member wingless-type MMTV integration site family member 16B (WNT16B). We determined that WNT16B expression is regulated by nuclear factor of κ light polypeptide gene enhancer in B cells 1 (NF-κB) after DNA damage and subsequently signals in a paracrine manner to activate the canonical Wnt program in tumor cells. The expression of WNT16B in the prostate tumor microenvironment attenuated the effects of cytotoxic chemotherapy in vivo, promoting tumor cell survival and disease progression. These results delineate a mechanism by which genotoxic therapies given in a cyclical manner can enhance subsequent treatment resistance through cell nonautonomous effects that are contributed by the tumor microenvironment.

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

Figure 1: Genotoxic damage to primary prostate fibroblasts induces expression of a spectrum of secreted proteins that includes WNT16B.
Figure 2: Cytotoxic chemotherapy induces WNT16B expression in the tumor microenvironment.
Figure 3: WNT16B is a major effector of the full DDSP and promotes the growth and invasion of prostate carcinoma.
Figure 4: Genotoxic stress upregulates WNT16B through NF-κB and signals through the canonical Wnt–β-catenin pathway to promote tumor cell proliferation and the acquisition of mesenchymal characteristics.
Figure 5: Paracrine-acting WNT16B promotes the resistance of prostate carcinoma to cytotoxic chemotherapy.
Figure 6: Chemotherapy resistance promoted by damaged fibroblasts is attenuated by blocking WNT16B, β-catenin or NF-κB signaling.

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Longley, D.B. & Johnston, P.G. Molecular mechanisms of drug resistance. J. Pathol. 205, 275–292 (2005).

    Article  CAS  Google Scholar 

  2. Wang, T.L. et al. Digital karyotyping identifies thymidylate synthase amplification as a mechanism of resistance to 5-fluorouracil in metastatic colorectal cancer patients. Proc. Natl. Acad. Sci. USA 101, 3089–3094 (2004).

    Article  CAS  Google Scholar 

  3. Schmitt, C.A., Rosenthal, C.T. & Lowe, S.W. Genetic analysis of chemoresistance in primary murine lymphomas. Nat. Med. 6, 1029–1035 (2000).

    Article  CAS  Google Scholar 

  4. Helmrich, A. et al. Recurrent chromosomal aberrations in INK4a/ARF defective primary lymphomas predict drug responses in vivo. Oncogene 24, 4174–4182 (2005).

    Article  CAS  Google Scholar 

  5. Redmond, K.M., Wilson, T.R., Johnston, P.G. & Longley, D.B. Resistance mechanisms to cancer chemotherapy. Front. Biosci. 13, 5138–5154 (2008).

    Article  CAS  Google Scholar 

  6. Wilson, T.R., Longley, D.B. & Johnston, P.G. Chemoresistance in solid tumours. Ann. Oncol. 17 (suppl. 10), x315–x324 (2006).

    Article  Google Scholar 

  7. Lee, S. & Schmitt, C.A. Chemotherapy response and resistance. Curr. Opin. Genet. Dev. 13, 90–96 (2003).

    Article  CAS  Google Scholar 

  8. Sakai, W. et al. Secondary mutations as a mechanism of cisplatin resistance in BRCA2-mutated cancers. Nature 451, 1116–1120 (2008).

    Article  CAS  Google Scholar 

  9. Kobayashi, H. et al. Acquired multicellular-mediated resistance to alkylating agents in cancer. Proc. Natl. Acad. Sci. USA 90, 3294–3298 (1993).

    Article  CAS  Google Scholar 

  10. Waldman, T. et al. Cell-cycle arrest versus cell death in cancer therapy. Nat. Med. 3, 1034–1036 (1997).

    Article  CAS  Google Scholar 

  11. Samson, D.J., Seidenfeld, J., Ziegler, K. & Aronson, N. Chemotherapy sensitivity and resistance assays: a systematic review. J. Clin. Oncol. 22, 3618–3630 (2004).

    Article  CAS  Google Scholar 

  12. Croix, B.S. et al. Reversal by hyaluronidase of adhesion-dependent multicellular drug resistance in mammary carcinoma cells. J. Natl. Cancer Inst. 88, 1285–1296 (1996).

    Article  CAS  Google Scholar 

  13. Kerbel, R.S. Molecular and physiologic mechanisms of drug resistance in cancer: an overview. Cancer Metastasis Rev. 20, 1–2 (2001).

    Article  CAS  Google Scholar 

  14. Wang, F. et al. Phenotypic reversion or death of cancer cells by altering signaling pathways in three-dimensional contexts. J. Natl. Cancer Inst. 94, 1494–1503 (2002).

    Article  CAS  Google Scholar 

  15. Kim, J.J. & Tannock, I.F. Repopulation of cancer cells during therapy: an important cause of treatment failure. Nat. Rev. Cancer 5, 516–525 (2005).

    Article  CAS  Google Scholar 

  16. Trédan, O., Galmarini, C.M., Patel, K. & Tannock, I.F. Drug resistance and the solid tumor microenvironment. J. Natl. Cancer Inst. 99, 1441–1454 (2007).

    Article  Google Scholar 

  17. Garzotto, M., Myrthue, A., Higano, C.S. & Beer, T.M. Neoadjuvant mitoxantrone and docetaxel for high-risk localized prostate cancer. Urol. Oncol. 24, 254–259 (2006).

    Article  CAS  Google Scholar 

  18. Beer, T.M. et al. Phase I study of weekly mitoxantrone and docetaxel before prostatectomy in patients with high-risk localized prostate cancer. Clin. Cancer Res. 10, 1306–1311 (2004).

    Article  CAS  Google Scholar 

  19. Bavik, C. et al. The gene expression program of prostate fibroblast senescence modulates neoplastic epithelial cell proliferation through paracrine mechanisms. Cancer Res. 66, 794–802 (2006).

    Article  CAS  Google Scholar 

  20. Coppé, J.P. et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS Biol. 6, 2853–2868 (2008).

    Article  Google Scholar 

  21. Clevers, H. Wnt/β-catenin signaling in development and disease. Cell 127, 469–480 (2006).

    Article  CAS  Google Scholar 

  22. Binet, R. et al. WNT16B is a new marker of cellular senescence that regulates p53 activity and the phosphoinositide 3-kinase/AKT pathway. Cancer Res. 69, 9183–9191 (2009).

    Article  CAS  Google Scholar 

  23. Acosta, J.C. et al. Chemokine signaling via the CXCR2 receptor reinforces senescence. Cell 133, 1006–1018 (2008).

    Article  CAS  Google Scholar 

  24. Huang, S.M. et al. Tankyrase inhibition stabilizes axin and antagonizes Wnt signalling. Nature 461, 614–620 (2009).

    Article  CAS  Google Scholar 

  25. Thiery, J.P., Acloque, H., Huang, R.Y. & Nieto, M.A. Epithelial-mesenchymal transitions in development and disease. Cell 139, 871–890 (2009).

    Article  CAS  Google Scholar 

  26. Yook, J.I. et al. A Wnt-Axin2–GSK3β cascade regulates Snail1 activity in breast cancer cells. Nat. Cell Biol. 8, 1398–1406 (2006).

    Article  CAS  Google Scholar 

  27. Vincan, E. & Barker, N. The upstream components of the Wnt signalling pathway in the dynamic EMT and MET associated with colorectal cancer progression. Clin. Exp. Metastasis 25, 657–663 (2008).

    Article  CAS  Google Scholar 

  28. Wu, K. & Bonavida, B. The activated NF-κB–Snail–RKIP circuitry in cancer regulates both the metastatic cascade and resistance to apoptosis by cytotoxic drugs. Crit. Rev. Immunol. 29, 241–254 (2009).

    Article  CAS  Google Scholar 

  29. Peinado, H., Olmeda, D. & Cano, A. Snail, Zeb and bHLH factors in tumour progression: an alliance against the epithelial phenotype? Nat. Rev. Cancer 7, 415–428 (2007).

    Article  CAS  Google Scholar 

  30. Bernard, D. et al. Involvement of Rel/nuclear factor-κB transcription factors in keratinocyte senescence. Cancer Res. 64, 472–481 (2004).

    Article  CAS  Google Scholar 

  31. Berchtold, C.M., Wu, Z.H., Huang, T.T. & Miyamoto, S. Calcium-dependent regulation of NEMO nuclear export in response to genotoxic stimuli. Mol. Cell. Biol. 27, 497–509 (2007).

    Article  CAS  Google Scholar 

  32. Kuilman, T. et al. Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network. Cell 133, 1019–1031 (2008).

    Article  CAS  Google Scholar 

  33. Kuilman, T. & Peeper, D.S. Senescence-messaging secretome: SMS-ing cellular stress. Nat. Rev. Cancer 9, 81–94 (2009).

    Article  CAS  Google Scholar 

  34. Fumagalli, M. & d'Adda di Fagagna, F. SASPense and DDRama in cancer and ageing. Nat. Cell Biol. 11, 921–923 (2009).

    Article  CAS  Google Scholar 

  35. Michaloglou, C. et al. BRAFE600-associated senescence-like cell cycle arrest of human naevi. Nature 436, 720–724 (2005).

    Article  CAS  Google Scholar 

  36. Xue, W. et al. Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas. Nature 445, 656–660 (2007).

    Article  CAS  Google Scholar 

  37. Wajapeyee, N., Serra, R.W., Zhu, X., Mahalingam, M. & Green, M.R. Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7. Cell 132, 363–374 (2008).

    Article  CAS  Google Scholar 

  38. Coppé, J.P., Kauser, K., Campisi, J. & Beausejour, C.M. Secretion of vascular endothelial growth factor by primary human fibroblasts at senescence. J. Biol. Chem. 281, 29568–29574 (2006).

    Article  Google Scholar 

  39. Gilbert, L.A. & Hemann, M.T. DNA damage–mediated induction of a chemoresistant niche. Cell 143, 355–366 (2010).

    Article  CAS  Google Scholar 

  40. Biswas, S. et al. Inhibition of TGF-β with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression. J. Clin. Invest. 117, 1305–1313 (2007).

    Article  CAS  Google Scholar 

  41. Davis, A.J. & Tannock, J.F. Repopulation of tumour cells between cycles of chemotherapy: a neglected factor. Lancet Oncol. 1, 86–93 (2000).

    Article  CAS  Google Scholar 

  42. Meads, M.B., Hazlehurst, L.A. & Dalton, W.S. The bone marrow microenvironment as a tumor sanctuary and contributor to drug resistance. Clin. Cancer Res. 14, 2519–2526 (2008).

    Article  CAS  Google Scholar 

  43. Shree, T. et al. Macrophages and cathepsin proteases blunt chemotherapeutic response in breast cancer. Genes Dev. 25, 2465–2479 (2011).

    Article  CAS  Google Scholar 

  44. DeNardo, D.G. et al. Leukocyte complexity predicts breast cancer survival and functionally regulates response to chemotherapy. Cancer Discov. 1, 54–67 (2011).

    Article  CAS  Google Scholar 

  45. Chien, Y. et al. Control of the senescence-associated secretory phenotype by NF-κB promotes senescence and enhances chemosensitivity. Genes Dev. 25, 2125–2136 (2011).

    Article  CAS  Google Scholar 

  46. Alderton, P.M., Gross, J. & Green, M.D. Comparative study of doxorubicin, mitoxantrone, and epirubicin in combination with ICRF-187 (ADR-529) in a chronic cardiotoxicity animal model. Cancer Res. 52, 194–201 (1992).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank J. Dean and D. Bianchi-Frias for helpful comments, A. Moreno for administrative assistance and N. Clegg for bioinformatics support. S. Hayward, Vanderbilt University, and J. Ware, Medical College of Virginia, provided BPH1 and M12 cells, respectively. Primary human prostate (PSC27), ovarian (OVF28901) and breast (HBF1203) fibroblasts were provided by B. Knudsen, Cedars Sinai Medical Center, E. Swisher, University of Washington, and P. Porter through the Seattle Breast SPORE (P50 CA138293), Fred Hutchinson Cancer Research Center, respectively. B. Torok-Strorb, Fred Hutchinson Cancer Research Center, provided HS5 and HS27A HPV E6/E7 immortalized human bone marrow stromal cells. We thank the clinicians who participated in the trials of neoadjuvant chemotherapy: M. Garzotto, T. Takayama, P. Lange, W. Ellis, S. Lieberman and B.A. Lowe. We are also grateful for the participation of the patients and their families in these studies. Breast cancer specimens were obtained from the Fred Hutchinson Cancer Research Center/University of Washington Medical Center Breast Specimen Repository. We thank N. Urban, Fred Hutchinson Cancer Research Center, for providing ovarian cancer biospecimens funded through the POCRC SPORE grant P50CA83636. This work was supported by a fellowship from the Department of Defense (PC073217), R01CA119125, the National Cancer Institute Tumor Microenvironment Network U54126540, the Pacific Northwest Prostate Cancer SPORE P50CA097186 and the Prostate Cancer Foundation.

Author information

Authors and Affiliations

Authors

Contributions

Y.S. designed and conducted experiments, and wrote the manuscript. J.C. provided reagents and technical advice. C.H., T.M.B. and P.P. provided clinical materials for the assessments of treatment responses. I.C. analyzed data. L.T. analyzed tissue histology and immunohistochemical assays. P.S.N. designed experiments, analyzed data and wrote the manuscript.

Corresponding author

Correspondence to Peter S Nelson.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Methods (PDF 10406 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sun, Y., Campisi, J., Higano, C. et al. Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat Med 18, 1359–1368 (2012). https://doi.org/10.1038/nm.2890

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.2890

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