Skip to main content
Erschienen in: Cellular Oncology 4/2019

17.04.2019 | Original Paper

Metformin enhances gefitinib efficacy by interfering with interactions between tumor-associated macrophages and head and neck squamous cell carcinoma cells

verfasst von: Xiteng Yin, Shengwei Han, Chuanhui Song, Huihui Zou, Zheng Wei, Wenguang Xu, Jianchuan Ran, Chuanchao Tang, Yufeng Wang, Yu Cai, Qingang Hu, Wei Han

Erschienen in: Cellular Oncology | Ausgabe 4/2019

Einloggen, um Zugang zu erhalten

Abstract

Background

Tumor-associated macrophages (TAMs) play an important role in drug resistance in many tumors, including head and neck squamous cell carcinoma (HNSCC). However, how TAMs interact with HNSCC cells to induce drug resistance, especially under hypoxic conditions, is unclear. In this study, we investigated the mechanism of TAM-induced gefitinib resistance in HNSCC cells and sought for novel therapeutic strategies.

Methods

The effects of hypoxia-treated HNSCC cells on the migration and polarization of macrophages were analyzed. Recombinant cytokine proteins and neutralizing antibodies were used as controls. In addition, we assessed the cytotoxic effects of gefitinib on HNSCC cells treated with M2-type macrophage conditioned medium, and carried out a cytokine antibody array analysis, thereby revealing the key factor CCL15. The relationship between serum CCL15 expression levels and prognosis in HNSCC patients was analyzed. In addition, we performed bioinformatic analyses to pursue the mechanisms of CCL15-induced gefitinib resistance. Finally, metformin was used to evaluate the sensitizing effects of gefitinib treatment on HNSCC cells in vitro and in vivo.

Results

We found that HNSCC cells recruited macrophages by secreting VEGF and polarized the macrophages to the M2 phenotype through IL-6. Conversely, we found that M2-type TAMs promoted HNSCC cell resistance to gefitinib through paracrine CCL15 signaling. The serum CCL15 levels in HNSCC patients showed a significant correlation with patient prognosis. Furthermore, we found that M2-type TAMs could suppress the sensitivity of HNSCC cells to gefitinib through the CCL15-CCR1-NF-κB pathway. In addition, we found that metformin not only inhibited CCL15 expression in M2-type TAMs enhanced by hypoxia, but also suppressed CCR1 surface expression in HNSCC cells. Encouragingly, we found that metformin sensitized HNSCC cells to gefitinib treatment in vitro and in vivo.

Conclusions

Based on our data we conclude that we have identified a novel interaction between M2-type TAMs and HNSCC cells that contributes to gefitinib resistance. We also found that metformin inhibited the cross-talk between macrophages and tumor cells, thereby eliciting therapeutic effects both in vitro and in vivo.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat M.J. Echarri, A. Lopez-Martin, R. Hitt, Targeted therapy in locally advanced and recurrent/metastatic head and neck squamous cell carcinoma (LA-R/M HNSCC). Cancers 8, 26 (2016) M.J. Echarri, A. Lopez-Martin, R. Hitt, Targeted therapy in locally advanced and recurrent/metastatic head and neck squamous cell carcinoma (LA-R/M HNSCC). Cancers 8, 26 (2016)
2.
Zurück zum Zitat S. Bhattacharyya, V. Sekar, B. Majumder, D.G. Mehrotra, S. Banerjee, A.K. Bhowmick, N. Alam, G.K. Mandal, J. Biswas, P.K. Majumder, N. Murmu, CDKN2A-p53 mediated antitumor effect of Lupeol in head and neck cancer. Cell Oncol 40, 145–155 (2017) S. Bhattacharyya, V. Sekar, B. Majumder, D.G. Mehrotra, S. Banerjee, A.K. Bhowmick, N. Alam, G.K. Mandal, J. Biswas, P.K. Majumder, N. Murmu, CDKN2A-p53 mediated antitumor effect of Lupeol in head and neck cancer. Cell Oncol 40, 145–155 (2017)
3.
Zurück zum Zitat C. Lo Nigro, N. Denaro, A. Merlotti, M. Merlano, Head and neck cancer: Improving outcomes with a multidisciplinary approach. Cancer Manag Res 9, 363–371 (2017)CrossRefPubMedPubMedCentral C. Lo Nigro, N. Denaro, A. Merlotti, M. Merlano, Head and neck cancer: Improving outcomes with a multidisciplinary approach. Cancer Manag Res 9, 363–371 (2017)CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat S. Roy, M. Kar, S. Roy, A. Saha, S. Padhi, B. Banerjee, Role of beta-catenin in cisplatin resistance, relapse and prognosis of head and neck squamous cell carcinoma. Cell Oncol 41, 185–200 (2018) S. Roy, M. Kar, S. Roy, A. Saha, S. Padhi, B. Banerjee, Role of beta-catenin in cisplatin resistance, relapse and prognosis of head and neck squamous cell carcinoma. Cell Oncol 41, 185–200 (2018)
5.
Zurück zum Zitat O. Casanovas, D.J. Hicklin, G. Bergers, D. Hanahan, Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 8, 299–309 (2005)CrossRefPubMed O. Casanovas, D.J. Hicklin, G. Bergers, D. Hanahan, Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 8, 299–309 (2005)CrossRefPubMed
6.
Zurück zum Zitat F. Shojaei, N. Ferrara, Role of the microenvironment in tumor growth and in refractoriness/resistance to anti-angiogenic therapies. Drug Resist Updat 11, 219–230 (2008)CrossRefPubMed F. Shojaei, N. Ferrara, Role of the microenvironment in tumor growth and in refractoriness/resistance to anti-angiogenic therapies. Drug Resist Updat 11, 219–230 (2008)CrossRefPubMed
7.
Zurück zum Zitat L.B. Rivera, G. Bergers, CANCER. Tumor angiogenesis, from foe to friend. Science 349, 694–695 (2015)CrossRefPubMed L.B. Rivera, G. Bergers, CANCER. Tumor angiogenesis, from foe to friend. Science 349, 694–695 (2015)CrossRefPubMed
8.
Zurück zum Zitat S.O. Lim, C.W. Li, W. Xia, H.H. Lee, S.S. Chang, J. Shen, J.L. Hsu, D. Raftery, D. Djukovic, H. Gu, W.C. Chang, H.L. Wang, M.L. Chen, L. Huo, C.H. Chen, Y. Wu, A. Sahin, S.M. Hanash, G.N. Hortobagyi, M.C. Hung, EGFR signaling enhances aerobic glycolysis in triple-negative breast Cancer cells to promote tumor growth and immune escape. Cancer Res 76, 1284–1296 (2016)CrossRefPubMedPubMedCentral S.O. Lim, C.W. Li, W. Xia, H.H. Lee, S.S. Chang, J. Shen, J.L. Hsu, D. Raftery, D. Djukovic, H. Gu, W.C. Chang, H.L. Wang, M.L. Chen, L. Huo, C.H. Chen, Y. Wu, A. Sahin, S.M. Hanash, G.N. Hortobagyi, M.C. Hung, EGFR signaling enhances aerobic glycolysis in triple-negative breast Cancer cells to promote tumor growth and immune escape. Cancer Res 76, 1284–1296 (2016)CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat C. Pozzi, A. Cuomo, I. Spadoni, E. Magni, A. Silvola, A. Conte, S. Sigismund, P.S. Ravenda, T. Bonaldi, M.G. Zampino, C. Cancelliere, P.P. Di Fiore, A. Bardelli, G. Penna, M. Rescigno, The EGFR-specific antibody cetuximab combined with chemotherapy triggers immunogenic cell death. Nat Med 22, 624–631 (2016)CrossRefPubMed C. Pozzi, A. Cuomo, I. Spadoni, E. Magni, A. Silvola, A. Conte, S. Sigismund, P.S. Ravenda, T. Bonaldi, M.G. Zampino, C. Cancelliere, P.P. Di Fiore, A. Bardelli, G. Penna, M. Rescigno, The EGFR-specific antibody cetuximab combined with chemotherapy triggers immunogenic cell death. Nat Med 22, 624–631 (2016)CrossRefPubMed
10.
Zurück zum Zitat S.N. Kazaz, I. Oztop, Treatment after first-generation epidermal growth factor receptor tyrosine kinase inhibitor resistance in non-small-cell lung Cancer. Turk Thorac J 18, 66–71 (2017)CrossRefPubMedPubMedCentral S.N. Kazaz, I. Oztop, Treatment after first-generation epidermal growth factor receptor tyrosine kinase inhibitor resistance in non-small-cell lung Cancer. Turk Thorac J 18, 66–71 (2017)CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat O. Tetsu, M.J. Hangauer, J. Phuchareon, D.W. Eisele, F. McCormick, Drug resistance to EGFR inhibitors in lung Cancer. Chemotherapy 61, 223–235 (2016)CrossRefPubMed O. Tetsu, M.J. Hangauer, J. Phuchareon, D.W. Eisele, F. McCormick, Drug resistance to EGFR inhibitors in lung Cancer. Chemotherapy 61, 223–235 (2016)CrossRefPubMed
12.
Zurück zum Zitat J.B. Cooper, E.E. Cohen, Mechanisms of resistance to EGFR inhibitors in head and neck cancer. Head Neck 31, 1086–1094 (2009)CrossRefPubMed J.B. Cooper, E.E. Cohen, Mechanisms of resistance to EGFR inhibitors in head and neck cancer. Head Neck 31, 1086–1094 (2009)CrossRefPubMed
13.
Zurück zum Zitat S. Ma, S. Pradeep, W. Hu, D. Zhang, R. Coleman, A. Sood, The role of tumor microenvironment in resistance to anti-angiogenic therapy. F1000Res 7, 326 (2018)CrossRefPubMedPubMedCentral S. Ma, S. Pradeep, W. Hu, D. Zhang, R. Coleman, A. Sood, The role of tumor microenvironment in resistance to anti-angiogenic therapy. F1000Res 7, 326 (2018)CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat M.C. Ocana, B. Martinez-Poveda, A.R. Quesada, M.A. Medina, Metabolism within the tumor microenvironment and its implication on cancer progression: An ongoing therapeutic target. Med Res Rev 39, 70-113 (2019) M.C. Ocana, B. Martinez-Poveda, A.R. Quesada, M.A. Medina, Metabolism within the tumor microenvironment and its implication on cancer progression: An ongoing therapeutic target. Med Res Rev 39, 70-113 (2019)
15.
Zurück zum Zitat S. Han, W. Xu, Z. Wang, X. Qi, Y. Wang, Y. Ni, H. Shen, Q. Hu, W. Han, Crosstalk between the HIF-1 and Toll-like receptor/nuclear factor-kappaB pathways in the oral squamous cell carcinoma microenvironment. Oncotarget 7, 37773–37789 (2016)PubMedPubMedCentral S. Han, W. Xu, Z. Wang, X. Qi, Y. Wang, Y. Ni, H. Shen, Q. Hu, W. Han, Crosstalk between the HIF-1 and Toll-like receptor/nuclear factor-kappaB pathways in the oral squamous cell carcinoma microenvironment. Oncotarget 7, 37773–37789 (2016)PubMedPubMedCentral
16.
Zurück zum Zitat X. Qi, W. Xu, J. Xie, Y. Wang, S. Han, Z. Wei, Y. Ni, Y. Dong, W. Han, Metformin sensitizes the response of oral squamous cell carcinoma to cisplatin treatment through inhibition of NF-kappaB/HIF-1alpha signal axis. Sci Rep 6, 35788 (2016)CrossRefPubMedPubMedCentral X. Qi, W. Xu, J. Xie, Y. Wang, S. Han, Z. Wei, Y. Ni, Y. Dong, W. Han, Metformin sensitizes the response of oral squamous cell carcinoma to cisplatin treatment through inhibition of NF-kappaB/HIF-1alpha signal axis. Sci Rep 6, 35788 (2016)CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat X. Yin, Z. Wei, C. Song, C. Tang, W. Xu, Y. Wang, J. Xie, Z. Lin, W. Han, Metformin sensitizes hypoxia-induced gefitinib treatment resistance of HNSCC via cell cycle regulation and EMT reversal. Cancer Manag Res 10, 5785–5798 (2018)CrossRefPubMedPubMedCentral X. Yin, Z. Wei, C. Song, C. Tang, W. Xu, Y. Wang, J. Xie, Z. Lin, W. Han, Metformin sensitizes hypoxia-induced gefitinib treatment resistance of HNSCC via cell cycle regulation and EMT reversal. Cancer Manag Res 10, 5785–5798 (2018)CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat A.C. Johansson, A. Ansell, F. Jerhammar, M.B. Lindh, R. Grenman, E. Munck-Wikland, A. Ostman, K. Roberg, Cancer-associated fibroblasts induce matrix metalloproteinase-mediated cetuximab resistance in head and neck squamous cell carcinoma cells. Mol Cancer Res 10, 1158–1168 (2012)CrossRefPubMed A.C. Johansson, A. Ansell, F. Jerhammar, M.B. Lindh, R. Grenman, E. Munck-Wikland, A. Ostman, K. Roberg, Cancer-associated fibroblasts induce matrix metalloproteinase-mediated cetuximab resistance in head and neck squamous cell carcinoma cells. Mol Cancer Res 10, 1158–1168 (2012)CrossRefPubMed
19.
20.
Zurück zum Zitat L.M. Nusblat, M.J. Carroll, C.M. Roth, Crosstalk between M2 macrophages and glioma stem cells. Cell Oncol 40, 471–482 (2017) L.M. Nusblat, M.J. Carroll, C.M. Roth, Crosstalk between M2 macrophages and glioma stem cells. Cell Oncol 40, 471–482 (2017)
21.
Zurück zum Zitat N. Eiro, L. Gonzalez, A. Martinez-Ordonez, B. Fernandez-Garcia, L.O. Gonzalez, S. Cid, F. Dominguez, R. Perez-Fernandez, F.J. Vizoso, Cancer-associated fibroblasts affect breast cancer cell gene expression, invasion and angiogenesis. Cell Oncol 41, 369–378 (2018) N. Eiro, L. Gonzalez, A. Martinez-Ordonez, B. Fernandez-Garcia, L.O. Gonzalez, S. Cid, F. Dominguez, R. Perez-Fernandez, F.J. Vizoso, Cancer-associated fibroblasts affect breast cancer cell gene expression, invasion and angiogenesis. Cell Oncol 41, 369–378 (2018)
22.
Zurück zum Zitat P. Nilendu, S.C. Sarode, D. Jahagirdar, I. Tandon, S. Patil, G.S. Sarode, J.K. Pal, N.K. Sharma, Mutual concessions and compromises between stromal cells and cancer cells: driving tumor development and drug resistance. Cell Oncol 41, 353–367 (2018) P. Nilendu, S.C. Sarode, D. Jahagirdar, I. Tandon, S. Patil, G.S. Sarode, J.K. Pal, N.K. Sharma, Mutual concessions and compromises between stromal cells and cancer cells: driving tumor development and drug resistance. Cell Oncol 41, 353–367 (2018)
23.
Zurück zum Zitat S. Stieb, A. Eleftheriou, G. Warnock, M. Guckenberger, O. Riesterer, Longitudinal PET imaging of tumor hypoxia during the course of radiotherapy. Eur J Nucl Med Mol Imaging 45, 2201–2217 (2018)CrossRefPubMed S. Stieb, A. Eleftheriou, G. Warnock, M. Guckenberger, O. Riesterer, Longitudinal PET imaging of tumor hypoxia during the course of radiotherapy. Eur J Nucl Med Mol Imaging 45, 2201–2217 (2018)CrossRefPubMed
24.
Zurück zum Zitat K. Harada, T. Ferdous, T. Harada, Y. Ueyama, Metformin in combination with 5-fluorouracil suppresses tumor growth by inhibiting the Warburg effect in human oral squamous cell carcinoma. Int J Oncol 49, 276–284 (2016)CrossRefPubMed K. Harada, T. Ferdous, T. Harada, Y. Ueyama, Metformin in combination with 5-fluorouracil suppresses tumor growth by inhibiting the Warburg effect in human oral squamous cell carcinoma. Int J Oncol 49, 276–284 (2016)CrossRefPubMed
25.
Zurück zum Zitat C. Murdoch, A. Giannoudis, C.E. Lewis, Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood 104, 2224–2234 (2004)CrossRefPubMed C. Murdoch, A. Giannoudis, C.E. Lewis, Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood 104, 2224–2234 (2004)CrossRefPubMed
26.
Zurück zum Zitat C. Tripathi, B.N. Tewari, R.K. Kanchan, K.S. Baghel, N. Nautiyal, R. Shrivastava, H. Kaur, M.L. Bhatt, S. Bhadauria, Macrophages are recruited to hypoxic tumor areas and acquire a pro-angiogenic M2-polarized phenotype via hypoxic cancer cell derived cytokines Oncostatin M and Eotaxin. Oncotarget 5, 5350–5368 (2014)CrossRefPubMedPubMedCentral C. Tripathi, B.N. Tewari, R.K. Kanchan, K.S. Baghel, N. Nautiyal, R. Shrivastava, H. Kaur, M.L. Bhatt, S. Bhadauria, Macrophages are recruited to hypoxic tumor areas and acquire a pro-angiogenic M2-polarized phenotype via hypoxic cancer cell derived cytokines Oncostatin M and Eotaxin. Oncotarget 5, 5350–5368 (2014)CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Y. Deng, Y. Yang, B. Yao, L. Ma, Q. Wu, Z. Yang, L. Zhang, B. Liu, Paracrine signaling by VEGF-C promotes non-small cell lung cancer cell metastasis via recruitment of tumor-associated macrophages. Exp Cell Res 364, 208–216 (2018)CrossRefPubMed Y. Deng, Y. Yang, B. Yao, L. Ma, Q. Wu, Z. Yang, L. Zhang, B. Liu, Paracrine signaling by VEGF-C promotes non-small cell lung cancer cell metastasis via recruitment of tumor-associated macrophages. Exp Cell Res 364, 208–216 (2018)CrossRefPubMed
28.
Zurück zum Zitat T. Chanmee, P. Ontong, K. Konno, N. Itano, Tumor-associated macrophages as major players in the tumor microenvironment. Cancers 6, 1670–1690 (2014) T. Chanmee, P. Ontong, K. Konno, N. Itano, Tumor-associated macrophages as major players in the tumor microenvironment. Cancers 6, 1670–1690 (2014)
29.
Zurück zum Zitat Z. Zhou, Y. Peng, X. Wu, S. Meng, W. Yu, J. Zhao, H. Zhang, J. Wang, W. Li, CCL18 secreted from M2 macrophages promotes migration and invasion via the PI3K/Akt pathway in gallbladder cancer. Cell Oncol 42, 81–92 (2019) Z. Zhou, Y. Peng, X. Wu, S. Meng, W. Yu, J. Zhao, H. Zhang, J. Wang, W. Li, CCL18 secreted from M2 macrophages promotes migration and invasion via the PI3K/Akt pathway in gallbladder cancer. Cell Oncol 42, 81–92 (2019)
30.
Zurück zum Zitat L. Ding, G. Liang, Z. Yao, J. Zhang, R. Liu, H. Chen, Y. Zhou, H. Wu, B. Yang, Q. He, Metformin prevents cancer metastasis by inhibiting M2-like polarization of tumor associated macrophages. Oncotarget 6, 36441–36455 (2015)PubMedPubMedCentral L. Ding, G. Liang, Z. Yao, J. Zhang, R. Liu, H. Chen, Y. Zhou, H. Wu, B. Yang, Q. He, Metformin prevents cancer metastasis by inhibiting M2-like polarization of tumor associated macrophages. Oncotarget 6, 36441–36455 (2015)PubMedPubMedCentral
31.
Zurück zum Zitat M.J. Schonenberger, W.J. Kovacs, Hypoxia signaling pathways: modulators of oxygen-related organelles. Front Cell Dev Biol 3(42) (2015) M.J. Schonenberger, W.J. Kovacs, Hypoxia signaling pathways: modulators of oxygen-related organelles. Front Cell Dev Biol 3(42) (2015)
32.
Zurück zum Zitat K.L. Talks, H. Turley, K.C. Gatter, P.H. Maxwell, C.W. Pugh, P.J. Ratcliffe, A.L. Harris, The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated macrophages. Am J Pathol 157, 411–421 (2000)CrossRefPubMedPubMedCentral K.L. Talks, H. Turley, K.C. Gatter, P.H. Maxwell, C.W. Pugh, P.J. Ratcliffe, A.L. Harris, The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated macrophages. Am J Pathol 157, 411–421 (2000)CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Y. Li, J. Wu, P. Zhang, CCL15/CCR1 axis is involved in hepatocellular carcinoma cells migration and invasion. Tumour Biol 37, 4501–4507 (2016)CrossRefPubMed Y. Li, J. Wu, P. Zhang, CCL15/CCR1 axis is involved in hepatocellular carcinoma cells migration and invasion. Tumour Biol 37, 4501–4507 (2016)CrossRefPubMed
34.
Zurück zum Zitat L.Z. Liu, Z. Zhang, B.H. Zheng, Y. Shi, M. Duan, L.J. Ma, Z.C. Wang, L.Q. Dong, P.P. Dong, J.Y. Shi, S. Zhang, Z.B. Ding, A.W. Ke, Y. Cao, X.M. Zhang, R. Xi, J. Zhou, J. Fan, X.Y. Wang, Q. Gao, CCL15 recruits suppressive monocytes to facilitate immune escape and disease progression in hepatocellular carcinoma. Hepatology 69, 143-159 (2019) L.Z. Liu, Z. Zhang, B.H. Zheng, Y. Shi, M. Duan, L.J. Ma, Z.C. Wang, L.Q. Dong, P.P. Dong, J.Y. Shi, S. Zhang, Z.B. Ding, A.W. Ke, Y. Cao, X.M. Zhang, R. Xi, J. Zhou, J. Fan, X.Y. Wang, Q. Gao, CCL15 recruits suppressive monocytes to facilitate immune escape and disease progression in hepatocellular carcinoma. Hepatology 69, 143-159 (2019)
35.
Zurück zum Zitat J.D. Qin, Z.H. Cao, X.F. Li, X.L. Kang, Y. Xue, Y.L. Li, D. Zhang, X.Y. Liu, Y.Z. Xue, Effect of ammonium pyrrolidine dithiocarbamate (PDTC) on NF-kappaB activation and CYP2E1 content of rats with immunological liver injury. Pharm Biol 52, 1460–1466 (2014)CrossRefPubMed J.D. Qin, Z.H. Cao, X.F. Li, X.L. Kang, Y. Xue, Y.L. Li, D. Zhang, X.Y. Liu, Y.Z. Xue, Effect of ammonium pyrrolidine dithiocarbamate (PDTC) on NF-kappaB activation and CYP2E1 content of rats with immunological liver injury. Pharm Biol 52, 1460–1466 (2014)CrossRefPubMed
36.
Zurück zum Zitat T. Kitamura, T. Fujishita, P. Loetscher, L. Revesz, H. Hashida, S. Kizaka-Kondoh, M. Aoki, M.M. Taketo, Inactivation of chemokine (C-C motif) receptor 1 (CCR1) suppresses colon cancer liver metastasis by blocking accumulation of immature myeloid cells in a mouse model. Proc Natl Acad Sci U S A 107, 13063–13068 (2010)CrossRefPubMedPubMedCentral T. Kitamura, T. Fujishita, P. Loetscher, L. Revesz, H. Hashida, S. Kizaka-Kondoh, M. Aoki, M.M. Taketo, Inactivation of chemokine (C-C motif) receptor 1 (CCR1) suppresses colon cancer liver metastasis by blocking accumulation of immature myeloid cells in a mouse model. Proc Natl Acad Sci U S A 107, 13063–13068 (2010)CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat A. Laitala, J.T. Erler, Hypoxic signalling in tumour stroma. Front Oncol 8, 189 (2018) A. Laitala, J.T. Erler, Hypoxic signalling in tumour stroma. Front Oncol 8, 189 (2018)
38.
Zurück zum Zitat B. Barleon, S. Sozzani, D. Zhou, H.A. Weich, A. Mantovani, D. Marme, Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1. Blood 87, 3336–3343 (1996)PubMed B. Barleon, S. Sozzani, D. Zhou, H.A. Weich, A. Mantovani, D. Marme, Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt-1. Blood 87, 3336–3343 (1996)PubMed
39.
Zurück zum Zitat L. Chen, S. Wang, Y. Wang, W. Zhang, K. Ma, C. Hu, H. Zhu, S. Liang, M. Liu, N. Xu, IL-6 influences the polarization of macrophages and the formation and growth of colorectal tumor. Oncotarget 9, 17443–17454 (2018)PubMedPubMedCentral L. Chen, S. Wang, Y. Wang, W. Zhang, K. Ma, C. Hu, H. Zhu, S. Liang, M. Liu, N. Xu, IL-6 influences the polarization of macrophages and the formation and growth of colorectal tumor. Oncotarget 9, 17443–17454 (2018)PubMedPubMedCentral
40.
Zurück zum Zitat A. Pardigol, U. Forssmann, H.D. Zucht, P. Loetscher, P. Schulz-Knappe, M. Baggiolini, W.G. Forssmann, H.J. Magert, HCC-2, a human chemokine: gene structure, expression pattern, and biological activity. Proc Natl Acad Sci U S A 95, 6308–6313 (1998)CrossRefPubMedPubMedCentral A. Pardigol, U. Forssmann, H.D. Zucht, P. Loetscher, P. Schulz-Knappe, M. Baggiolini, W.G. Forssmann, H.J. Magert, HCC-2, a human chemokine: gene structure, expression pattern, and biological activity. Proc Natl Acad Sci U S A 95, 6308–6313 (1998)CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Y. Li, H.P. Yu, P. Zhang, CCL15 overexpression predicts poor prognosis for hepatocellular carcinoma. Hepatol Int 10, 488–492 (2016)CrossRefPubMed Y. Li, H.P. Yu, P. Zhang, CCL15 overexpression predicts poor prognosis for hepatocellular carcinoma. Hepatol Int 10, 488–492 (2016)CrossRefPubMed
42.
Zurück zum Zitat N.J. Beasley, R. Leek, M. Alam, H. Turley, G.J. Cox, K. Gatter, P. Millard, S. Fuggle, A.L. Harris, Hypoxia-inducible factors HIF-1alpha and HIF-2alpha in head and neck cancer: relationship to tumor biology and treatment outcome in surgically resected patients. Cancer Res 62, 2493–2497 (2002)PubMed N.J. Beasley, R. Leek, M. Alam, H. Turley, G.J. Cox, K. Gatter, P. Millard, S. Fuggle, A.L. Harris, Hypoxia-inducible factors HIF-1alpha and HIF-2alpha in head and neck cancer: relationship to tumor biology and treatment outcome in surgically resected patients. Cancer Res 62, 2493–2497 (2002)PubMed
43.
Zurück zum Zitat X.S. Deng, S. Wang, A. Deng, B. Liu, S.M. Edgerton, S.E. Lind, R. Wahdan-Alaswad, A.D. Thor, Metformin targets Stat3 to inhibit cell growth and induce apoptosis in triple-negative breast cancers. Cell Cycle 11, 367–376 (2012)CrossRefPubMed X.S. Deng, S. Wang, A. Deng, B. Liu, S.M. Edgerton, S.E. Lind, R. Wahdan-Alaswad, A.D. Thor, Metformin targets Stat3 to inhibit cell growth and induce apoptosis in triple-negative breast cancers. Cell Cycle 11, 367–376 (2012)CrossRefPubMed
44.
Zurück zum Zitat L. Li, R. Han, H. Xiao, C. Lin, Y. Wang, H. Liu, K. Li, H. Chen, F. Sun, Z. Yang, J. Jiang, Y. He, Metformin sensitizes EGFR-TKI-resistant human lung cancer cells in vitro and in vivo through inhibition of IL-6 signaling and EMT reversal. Clin Cancer Res 20, 2714–2726 (2014)CrossRefPubMed L. Li, R. Han, H. Xiao, C. Lin, Y. Wang, H. Liu, K. Li, H. Chen, F. Sun, Z. Yang, J. Jiang, Y. He, Metformin sensitizes EGFR-TKI-resistant human lung cancer cells in vitro and in vivo through inhibition of IL-6 signaling and EMT reversal. Clin Cancer Res 20, 2714–2726 (2014)CrossRefPubMed
45.
Zurück zum Zitat S. Eikawa, M. Nishida, S. Mizukami, C. Yamazaki, E. Nakayama, H. Udono, Immune-mediated antitumor effect by type 2 diabetes drug, metformin. Proc Natl Acad Sci U S A 112, 1809–1814 (2015)CrossRefPubMedPubMedCentral S. Eikawa, M. Nishida, S. Mizukami, C. Yamazaki, E. Nakayama, H. Udono, Immune-mediated antitumor effect by type 2 diabetes drug, metformin. Proc Natl Acad Sci U S A 112, 1809–1814 (2015)CrossRefPubMedPubMedCentral
Metadaten
Titel
Metformin enhances gefitinib efficacy by interfering with interactions between tumor-associated macrophages and head and neck squamous cell carcinoma cells
verfasst von
Xiteng Yin
Shengwei Han
Chuanhui Song
Huihui Zou
Zheng Wei
Wenguang Xu
Jianchuan Ran
Chuanchao Tang
Yufeng Wang
Yu Cai
Qingang Hu
Wei Han
Publikationsdatum
17.04.2019
Verlag
Springer Netherlands
Erschienen in
Cellular Oncology / Ausgabe 4/2019
Print ISSN: 2211-3428
Elektronische ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-019-00446-y

Weitere Artikel der Ausgabe 4/2019

Cellular Oncology 4/2019 Zur Ausgabe

Neu im Fachgebiet Pathologie

Molekularpathologische Untersuchungen im Wandel der Zeit

Open Access Biomarker Leitthema

Um auch an kleinen Gewebeproben zuverlässige und reproduzierbare Ergebnisse zu gewährleisten ist eine strenge Qualitätskontrolle in jedem Schritt des Arbeitsablaufs erforderlich. Eine nicht ordnungsgemäße Prüfung oder Behandlung des …

Vergleichende Pathologie in der onkologischen Forschung

Pathologie Leitthema

Die vergleichende experimentelle Pathologie („comparative experimental pathology“) ist ein Fachbereich an der Schnittstelle von Human- und Veterinärmedizin. Sie widmet sich der vergleichenden Erforschung von Gemeinsamkeiten und Unterschieden von …

Gastrointestinale Stromatumoren

Open Access GIST CME-Artikel

Gastrointestinale Stromatumoren (GIST) stellen seit über 20 Jahren ein Paradigma für die zielgerichtete Therapie mit Tyrosinkinaseinhibitoren dar. Eine elementare Voraussetzung für eine mögliche neoadjuvante oder adjuvante Behandlung bei …

Personalisierte Medizin in der Onkologie

Aufgrund des erheblichen technologischen Fortschritts in der molekularen und genetischen Diagnostik sowie zunehmender Erkenntnisse über die molekulare Pathogenese von Krankheiten hat in den letzten zwei Jahrzehnten ein grundlegender …