Skip to main content

Advertisement

Log in

Interaction between cancer cells and cancer-associated fibroblasts after cisplatin treatment promotes cancer cell regrowth

  • Research Article
  • Published:
Human Cell Aims and scope Submit manuscript

Abstract

Regrowth of cancer cells following chemotherapy is a significant problem for cancer patients. This study examined whether cancer-associated fibroblasts (CAFs), a major component of a tumor microenvironment, promote cancer cell regrowth after chemotherapy. First, we treated human lung adenocarcinoma cell line A549 and CAFs from four patients with cisplatin. Cisplatin treatment inhibited the viable cell number of A549 cells and induced epithelial–mesenchymal transition. After cisplatin was removed, A549 cells continued to manifest the mesenchymal phenotype and proliferated 2.2-fold in 4 days (regrowth of A549 cells). Cisplatin treatment inhibited the viable cell number of CAFs from four patients also. The CM (derived from cisplatin-pretreated CAFs from two patients) significantly enhanced the regrowth of cisplatin-pretreated A549 cells, and the CM derived from cisplatin-naïve CAFs marginally enhanced A549 regrowth. By contrast, the CM derived from either cisplatin-pretreated CAFs or cisplatin-naïve CAFs failed to enhance the growth of cisplatin-naïve A549 cells. The CM derived from cisplatin-pretreated CAFs did not enhance the proliferation of A549 cells in which epithelial–mesenchymal transition was induced by TGFβ-1. Our findings indicate the possibility that humoral factors from cisplatin-pretreated CAFs promote the regrowth of cisplatin-pretreated A549 cells. These results suggest that interactions between cancer cells and CAFs may significantly enhance cancer cell regrowth within the tumor microenvironment after cisplatin treatment.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Kaur A, Webster MR, Marchbank K, et al. sFRP2 in the aged microenvironment drives melanoma metastasis and therapy resistance. Nature. 2016;532:250–4.

    Article  CAS  Google Scholar 

  2. Patel GK, Khan MA, Bhardwaj A, et al. Exosomes confer chemoresistance to pancreatic cancer cells by promoting ROS detoxification and miR-155-mediated suppression of key gemcitabine-metabolising enzyme, DCK. Br J Cancer. 2017;116:609–19.

    Article  CAS  Google Scholar 

  3. Zhou W, Sun W, Yung MMH, et al. Autocrine activation of JAK2 by IL-11 promotes platinum drug resistance. Oncogene. 2018;37:3981–97.

    Article  CAS  Google Scholar 

  4. Aoyama A, Katayama R, Oh-Hara T, Sato S, Okuno Y, Fujita N. Tivantinib (ARQ 197) exhibits antitumor activity by directly interacting with tubulin and overcomes ABC transporter-mediated drug resistance. Mol Cancer Ther. 2014;13:2978–90.

    Article  CAS  Google Scholar 

  5. Katayama R, Kobayashi Y, Friboulet L, et al. Cabozantinib overcomes crizotinib resistance in ROS1 fusion-positive cancer. Clin Cancer Res. 2015;21:166–74.

    Article  CAS  Google Scholar 

  6. Zhou N, Wu X, Yang B, Yang X, Zhang D, Qing G. Stem cell characteristics of dormant cells and cisplatin-induced effects on the stemness of epithelial ovarian cancer cells. Mol Med Rep. 2014;10:2495–504.

    Article  CAS  Google Scholar 

  7. Galluzzi L, Senovilla L, Vitale I, et al. Molecular mechanisms of cisplatin resistance. Oncogene. 2012;31:1869–83.

    Article  CAS  Google Scholar 

  8. Liang SQ, Marti TM, Dorn P, et al. Blocking the epithelial-to-mesenchymal transition pathway abrogates resistance to anti-folate chemotherapy in lung cancer. Cell Death Dis. 2015;6:e1824.

    Article  CAS  Google Scholar 

  9. Whatcott Clifford J, Han Haiyong, Posner Richard G, Von Hoff Daniel D. Tumor-stromal interactions in pancreatic cancer. Crit Rev Oncog. 2013;18:135–51.

    Article  Google Scholar 

  10. Yashiro M, Hirakawa K. Cancer-stromal interactions in scirrhous gastric carcinoma. Cancer Microenviron. 2010;3:127–35.

    Article  CAS  Google Scholar 

  11. Hida K, Maishi N, Sakurai Y, Hida Y, Harashima H. Heterogeneity of tumor endothelial cells and drug delivery. Adv Drug Deliv Rev. 2016;99:140–7.

    Article  CAS  Google Scholar 

  12. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer. 2006;6:392–401.

    Article  CAS  Google Scholar 

  13. Hoshino A, Ishii G, Ito T, et al. Podoplanin-positive fibroblasts enhance lung adenocarcinoma tumor formation: podoplanin in fibroblast functions for tumor progression. Cancer Res. 2011;71:4769–79.

    Article  CAS  Google Scholar 

  14. Ishii G, Ochiai A, Neri S. Phenotypic and functional heterogeneity of cancer-associated fibroblast within the tumor microenvironment. Adv Drug Deliv Rev. 2016;99:186–96.

    Article  CAS  Google Scholar 

  15. Neri S, Ishii G, Hashimoto H, et al. Podoplanin-expressing cancer-associated fibroblasts lead and enhance the local invasion of cancer cells in lung adenocarcinoma. Int J Cancer. 2015;137:784–96.

    Article  CAS  Google Scholar 

  16. Fang T, Lv H, Lv G, et al. Tumor-derived exosomal miR-1247-3p induces cancer-associated fibroblast activation to foster lung metastasis of liver cancer. Nat Commun. 2018;9:191.

    Article  Google Scholar 

  17. Ishibashi M, Neri S, Hashimoto H, et al. CD200-positive cancer associated fibroblasts augment the sensitivity of Epidermal Growth Factor Receptor mutation-positive lung adenocarcinomas to EGFR Tyrosine kinase inhibitors. Sci Rep. 2017;7:46662.

    Article  Google Scholar 

  18. Roswall P, Bocci M, Bartoschek M, et al. Microenvironmental control of breast cancer subtype elicited through paracrine platelet-derived growth factor-CC signaling. Nat Med. 2018;24:463–73.

    Article  CAS  Google Scholar 

  19. Wang W, Li Q, Yamada T, et al. Crosstalk to stromal fibroblasts induces resistance of lung cancer to epidermal growth factor receptor tyrosine kinase inhibitors. Clin Cancer Res. 2009;15:6630–8.

    Article  CAS  Google Scholar 

  20. Yoshida T, Ishii G, Goto K, et al. Podoplanin-positive cancer-associated fibroblasts in the tumor microenvironment induce primary resistance to EGFR-TKIs in lung adenocarcinoma with EGFR mutation. Clin Cancer Res. 2015;21:642–51.

    Article  CAS  Google Scholar 

  21. Yang L, Fang J, Chen J. Tumor cell senescence response produces aggressive variants. Cell Death Discov. 2017;3:17049.

    Article  CAS  Google Scholar 

  22. Hashimoto H, Suda Y, Miyashita T, et al. A novel method to generate single-cell-derived cancer-associated fibroblast clones. J Cancer Res Clin Oncol. 2017;143:1409–19.

    Article  CAS  Google Scholar 

  23. Neri S, Miyashita T, Hashimoto H, et al. Fibroblast-led cancer cell invasion is activated by epithelial-mesenchymal transition through platelet-derived growth factor BB secretion of lung adenocarcinoma. Cancer Lett. 2017;395:20–30.

    Article  CAS  Google Scholar 

  24. Sun Y, Campisi J, Higano C, et al. Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat Med. 2012;18:1359–68.

    Article  CAS  Google Scholar 

  25. Lotti F, Jarrar AM, Pai RK, et al. Chemotherapy activates cancer-associated fibroblasts to maintain colorectal cancer-initiating cells by IL-17A. J Exp Med. 2013;210:2851–72.

    Article  CAS  Google Scholar 

  26. Tao L, Huang G, Wang R, et al. Cancer-associated fibroblasts treated with cisplatin facilitates chemoresistance of lung adenocarcinoma through IL-11/IL-11R/STAT3 signaling pathway. Sci Rep. 2016;6:38408.

    Article  CAS  Google Scholar 

  27. Chen QY, Jiao DM, Wang J, Hu H, Tang X, Chen J, Mou H, Lu W. miR-206 regulates cisplatin resistance and EMT in human lung adenocarcinoma cells partly by targeting MET. Oncotarget. 2016;7:24510–26.

    PubMed  PubMed Central  Google Scholar 

  28. Shintani Y, Okimura A, Sato K, et al. Epithelial to mesenchymal transition is a determinant of sensitivity to chemoradiotherapy in non-small cell lung cancer. Ann Thorac Surg. 2011;92:1794–804.

    Article  Google Scholar 

Download references

Funding

This work was supported in part by JSPS KAKENHI (16H05311).

Author information

Authors and Affiliations

Authors

Contributions

SH contributed to the design and coordination of the study, performed experiment, and prepared the manuscript. TM and HH performed the experiment, and read and approved the final manuscript. SN, MS, HN, SY, AO, KG and MT contributed to preparing the manuscript, and read and approved the final manuscript. GI contributed to the design and coordination of the study, revised the article for important intellectual content, and read and approved the final manuscript.

Corresponding author

Correspondence to Genichiro Ishii.

Ethics declarations

Conflict of interest

All authors declare that they have no conflict of interest.

Ethical approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards.

Informed consent

Comprehensive informed consent was obtained from all individual participants included in the study.

Additional information

Publisher's Note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Figure S1. Schematic of the main experiment. A) The experimental schematic of the effect of CM derived from CAFs treated with cisplatin on the growth of cisplatin-pretreated A549 cells. B) Experimental schematic of the effect of CM derived from naïve CAFs on the growth of naïve A549 cells.

Figure S2. Expression levels of 9 EMT related factors in cisplatin-pretreated A549 cells for 4 d following cisplatin removal. Values are means ± S.D. from three independent experiments. *p< 0.05

Figure S3. Expression levels of IL-6, HGF, VEGF-A, FGF-2, and IGF-1mRNA in naïve CAFs and cisplatin-treated CAFs. A) Expression levels of IL-6, HGF, VEGF-A, FGF-2, and IGF-1mRNA in naïve CAFs. Naïve and cisplatin-treated CAF1 and CAF3 don’t have promoting effect on A549 cell regrowth, while CAF2 and CAF4 have promoting effect. (Figure 3). First we examined expression levels of IL-6, HGF, VEGF-A, FGF-2, and IGF-1mRNA in naïve CAFs1,2,3, and 4 by RT-PCR method. The expression of HGF was obviously lower in naïve CAF2 and CAF4 compared to CAF1 and CAF3. B) Expression levels of IL-6, HGF, VEGF-A, FGF-2, and IGF-1mRNA in cisplatin-treated CAFs. HGF expression level in cisplatin-treated CAF2 was obviously higher than cisplatin-treated CAF1, which was the reverse result using naïve CAF1 and CAF2.

Figure S4. Summary of this study. A: In TME without cisplatin, CAFs did not increase the proliferation of A549 cells. B: In the TME following cisplatin treatment, cisplatin-exposed CAFs stimulated the growth of cisplatin-exposed A549 cells.

Supplementary material 1 (PPTX 271 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hisamitsu, S., Miyashita, T., Hashimoto, H. et al. Interaction between cancer cells and cancer-associated fibroblasts after cisplatin treatment promotes cancer cell regrowth. Human Cell 32, 453–464 (2019). https://doi.org/10.1007/s13577-019-00275-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s13577-019-00275-z

Keywords

Navigation