Skip to main content
Erschienen in: Cellular Oncology 1/2020

06.09.2019 | Original paper

Anti-tumor activity of cediranib, a pan-vascular endothelial growth factor receptor inhibitor, in pancreatic ductal adenocarcinoma cells

verfasst von: Majid Momeny, Zivar Alishahi, Haniyeh Eyvani, Fatemeh Esmaeili, Azam Zaghal, Parisa Ghaffari, Javad Tavakkoly-Bazzaz, Kamran Alimoghaddam, Ardeshir Ghavamzadeh, Seyed H. Ghaffari

Erschienen in: Cellular Oncology | Ausgabe 1/2020

Einloggen, um Zugang zu erhalten

Abstract

Purpose

Pancreatic ductal adenocarcinoma (PDAC) is the most common and lethal subtype of pancreatic cancer, with a 5-year survival rate of < 3%. Early tumor dissemination, late diagnosis and insensitivity to conventional treatment are the major reasons for its high mortality rate. Members of the vascular endothelial growth factor (VEGF) family are overexpressed in PDAC and play important roles in its malignant progression, suggesting that VEGF-targeted therapies may interrupt the proliferation and motility of PDAC cells. Here, we evaluated the anti-tumor activity of cediranib, a pan-VEGF receptor inhibitor, on PDAC cells.

Methods

Anti-proliferative effects of cediranib were determined using cell proliferation and crystal violet staining assays. Annexin V/PI staining, radiation therapy, and cell migration and invasion assays were carried out to examine the effects of cediranib on apoptosis, radio-sensitivity and cell motility, respectively. Quantitative reverse transcription-PCR (qRT-PCR) and Western blot analyses were applied to elucidate the molecular mechanisms underlying the anti-tumor activity of cediranib.

Results

We found that cediranib decreased PDAC cell proliferation and clonogenic survival and induced apoptotic cell death through inhibition of the anti-apoptotic proteins cIAP1, XIAP, MCL-1 and survivin. Combination with cediranib synergistically increased the sensitivity of PDAC cells to chemotherapeutic agents such as gemcitabine and paclitaxel, and potentiated the effects of radiation therapy on PDAC cell growth inhibition and apoptosis induction. Furthermore, we found that treatment with cediranib impaired PDAC cell migration and invasion via expression reduction of the epithelial-to-mesenchymal transition (EMT) markers ZEB1, N-cadherin and Snail.

Conclusions

Our data indicate that cediranib may exhibit anti-tumor activity in PDAC cells and provide a rationale for further investigation of the potential of VEGF receptor-targeted therapies for the treatment of PDAC.
Anhänge
Nur mit Berechtigung zugänglich
Literatur
1.
Zurück zum Zitat C.S. Yabar, J.M. Winter, Pancreatic cancer: A review. Gastroenterol. Clin. North Am. 45, 429–445 (2016)PubMedCrossRef C.S. Yabar, J.M. Winter, Pancreatic cancer: A review. Gastroenterol. Clin. North Am. 45, 429–445 (2016)PubMedCrossRef
2.
Zurück zum Zitat L. Rahib, B.D. Smith, R. Aizenberg, A.B. Rosenzweig, J.M. Fleshman, L.M. Matrisian, Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 74, 2913–2921 (2014)PubMedCrossRef L. Rahib, B.D. Smith, R. Aizenberg, A.B. Rosenzweig, J.M. Fleshman, L.M. Matrisian, Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 74, 2913–2921 (2014)PubMedCrossRef
3.
Zurück zum Zitat T. Muniraj, P.A. Jamidar, H.R. Aslanian, Pancreatic cancer: a comprehensive review and update. Dis. Mon. 11, 368–402 (2013)CrossRef T. Muniraj, P.A. Jamidar, H.R. Aslanian, Pancreatic cancer: a comprehensive review and update. Dis. Mon. 11, 368–402 (2013)CrossRef
4.
Zurück zum Zitat R.L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, 2018. CA Cancer J. Clin. 68, 7–30 (2018) R.L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, 2018. CA Cancer J. Clin. 68, 7–30 (2018)
5.
Zurück zum Zitat M. Chiaravalli, M. Reni, E.M. O'Reilly, Pancreatic ductal adenocarcinoma: State-of-the-art 2017 and new therapeutic strategies. Cancer Treat. Rev. 60, 32–43 (2017)PubMedCrossRef M. Chiaravalli, M. Reni, E.M. O'Reilly, Pancreatic ductal adenocarcinoma: State-of-the-art 2017 and new therapeutic strategies. Cancer Treat. Rev. 60, 32–43 (2017)PubMedCrossRef
6.
Zurück zum Zitat J. Kleeff, M. Korc, M. Apte, C. La Vecchia, C.D. Johnson, A.V. Biankin, R.E. Neale, M. Tempero, D.A. Tuveson, R.H. Hruban, Pancreatic cancer. Nat. Rev. Dis. Primers 2, 16022 (2016) J. Kleeff, M. Korc, M. Apte, C. La Vecchia, C.D. Johnson, A.V. Biankin, R.E. Neale, M. Tempero, D.A. Tuveson, R.H. Hruban, Pancreatic cancer. Nat. Rev. Dis. Primers 2, 16022 (2016)
7.
Zurück zum Zitat D. Singh, G. Upadhyay, R.K. Srivastava, S. Shankar, Recent advances in pancreatic cancer: biology, treatment, and prevention. Biochim. Biophys. Acta 1856, 13–27 (2015)CrossRef D. Singh, G. Upadhyay, R.K. Srivastava, S. Shankar, Recent advances in pancreatic cancer: biology, treatment, and prevention. Biochim. Biophys. Acta 1856, 13–27 (2015)CrossRef
8.
Zurück zum Zitat K.E. Craven, J. Gore, M. Korc, Overview of pre-clinical and clinical studies targeting angiogenesis in pancreatic ductal adenocarcinoma. Cancer Lett. 381, 201–210 (2016)PubMedCrossRef K.E. Craven, J. Gore, M. Korc, Overview of pre-clinical and clinical studies targeting angiogenesis in pancreatic ductal adenocarcinoma. Cancer Lett. 381, 201–210 (2016)PubMedCrossRef
9.
Zurück zum Zitat G. Goel, W. Sun, Novel approaches in the management of pancreatic ductal adenocarcinoma: potential promises for the future. J. Hematol. Oncol. 8, 44 (2015)PubMedPubMedCentralCrossRef G. Goel, W. Sun, Novel approaches in the management of pancreatic ductal adenocarcinoma: potential promises for the future. J. Hematol. Oncol. 8, 44 (2015)PubMedPubMedCentralCrossRef
11.
Zurück zum Zitat B. Al-Husein, M. Abdalla, M. Trepte, D.L. DeRemer, P.R. Somanath, Antiangiogenic therapy for cancer: an update. Pharmacotherapy 32, 1095–1111 (2012)PubMedPubMedCentralCrossRef B. Al-Husein, M. Abdalla, M. Trepte, D.L. DeRemer, P.R. Somanath, Antiangiogenic therapy for cancer: an update. Pharmacotherapy 32, 1095–1111 (2012)PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat A. Hoeben, B. Landuyt, M.S. Highley, H. Wildiers, A.T. Van Oosterom, E.A. De Bruijn, Vascular endothelial growth factor and angiogenesis. Pharmacol. Rev. 56, 549–580 (2004)PubMedCrossRef A. Hoeben, B. Landuyt, M.S. Highley, H. Wildiers, A.T. Van Oosterom, E.A. De Bruijn, Vascular endothelial growth factor and angiogenesis. Pharmacol. Rev. 56, 549–580 (2004)PubMedCrossRef
14.
Zurück zum Zitat J.-L. Su, P.-C. Yang, J.-Y. Shih, C.-Y. Yang, L.-H. Wei, C.-Y. Hsieh, C.-H. Chou, Y.-M. Jeng, M.-Y. Wang, K.-J. Chang, The VEGF-C/Flt-4 axis promotes invasion and metastasis of cancer cells. Cancer Cell 9, 209–223 (2006)PubMedCrossRef J.-L. Su, P.-C. Yang, J.-Y. Shih, C.-Y. Yang, L.-H. Wei, C.-Y. Hsieh, C.-H. Chou, Y.-M. Jeng, M.-Y. Wang, K.-J. Chang, The VEGF-C/Flt-4 axis promotes invasion and metastasis of cancer cells. Cancer Cell 9, 209–223 (2006)PubMedCrossRef
15.
Zurück zum Zitat R. Aesoy, B.C. Sanchez, J.H. Norum, R. Lewensohn, K. Viktorsson, B. Linderholm, An autocrine VEGF/VEGFR2 and p38 signaling loop confers resistance to 4-hydroxytamoxifen in MCF-7 breast cancer cells. Mol. Cancer Res. 6, 1630–1638 (2008)PubMedCrossRef R. Aesoy, B.C. Sanchez, J.H. Norum, R. Lewensohn, K. Viktorsson, B. Linderholm, An autocrine VEGF/VEGFR2 and p38 signaling loop confers resistance to 4-hydroxytamoxifen in MCF-7 breast cancer cells. Mol. Cancer Res. 6, 1630–1638 (2008)PubMedCrossRef
16.
Zurück zum Zitat J.-L. Su, C. Yen, P. Chen, S. Chuang, C. Hong, I. Kuo, H. Chen, M.-C. Hung, M. Kuo, The role of the VEGF-C/VEGFR-3 axis in cancer progression. Br. J. Cancer 96, 541 (2007)PubMedPubMedCentralCrossRef J.-L. Su, C. Yen, P. Chen, S. Chuang, C. Hong, I. Kuo, H. Chen, M.-C. Hung, M. Kuo, The role of the VEGF-C/VEGFR-3 axis in cancer progression. Br. J. Cancer 96, 541 (2007)PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Z. Von Marschall, T. Cramer, M. Höcker, R. Burde, T. Plath, M. Schirner, R. Heidenreich, G. Breier, E.O. Riecken, B. Wiedenmann, De novo expression of vascular endothelial growth factor in human pancreatic cancer: Evidence for an autocrine mitogenic loop. Gastroenterology 119, 1358–1372 (2000)CrossRef Z. Von Marschall, T. Cramer, M. Höcker, R. Burde, T. Plath, M. Schirner, R. Heidenreich, G. Breier, E.O. Riecken, B. Wiedenmann, De novo expression of vascular endothelial growth factor in human pancreatic cancer: Evidence for an autocrine mitogenic loop. Gastroenterology 119, 1358–1372 (2000)CrossRef
18.
Zurück zum Zitat M. Costache, M. Ioana, S. Iordache, D. Ene, C.A. Costache, A. Săftoiu, VEGF expression in pancreatic cancer and other malignancies: a review of the literature. Rom. J. Intern. Med. 53, 199–208 (2015)PubMed M. Costache, M. Ioana, S. Iordache, D. Ene, C.A. Costache, A. Săftoiu, VEGF expression in pancreatic cancer and other malignancies: a review of the literature. Rom. J. Intern. Med. 53, 199–208 (2015)PubMed
19.
Zurück zum Zitat A.D. Yang, E.R. Camp, F. Fan, L. Shen, M.J. Gray, W. Liu, R. Somcio, T.W. Bauer, Y. Wu, D.J. Hicklin, Vascular endothelial growth factor receptor-1 activation mediates epithelial to mesenchymal transition in human pancreatic carcinoma cells. Cancer Res. 66, 46–51 (2006)PubMedCrossRef A.D. Yang, E.R. Camp, F. Fan, L. Shen, M.J. Gray, W. Liu, R. Somcio, T.W. Bauer, Y. Wu, D.J. Hicklin, Vascular endothelial growth factor receptor-1 activation mediates epithelial to mesenchymal transition in human pancreatic carcinoma cells. Cancer Res. 66, 46–51 (2006)PubMedCrossRef
20.
Zurück zum Zitat J.P. Thiery, Epithelial–mesenchymal transitions in tumour progression. Nat. Rev. Cancer 2, 442 (2002)PubMedCrossRef J.P. Thiery, Epithelial–mesenchymal transitions in tumour progression. Nat. Rev. Cancer 2, 442 (2002)PubMedCrossRef
21.
Zurück zum Zitat Y. Doi, M. Yashiro, N. Yamada, R. Amano, S. Noda, K. Hirakawa, VEGF-A/VEGFR-2 signaling plays an important role for the motility of pancreas cancer cells. Ann. Surg. Oncol. 19, 2733–2743 (2012)PubMedCrossRef Y. Doi, M. Yashiro, N. Yamada, R. Amano, S. Noda, K. Hirakawa, VEGF-A/VEGFR-2 signaling plays an important role for the motility of pancreas cancer cells. Ann. Surg. Oncol. 19, 2733–2743 (2012)PubMedCrossRef
22.
Zurück zum Zitat Y. Doi, M. Yashiro, N. Yamada, R. Amano, G. Ohira, M. Komoto, S. Noda, S. Kashiwagi, Y. Kato, Y. Fuyuhiro, Significance of phospho-vascular endothelial growth factor receptor-2 expression in pancreatic cancer. Cancer Sci. 101, 1529–1535 (2010)PubMedCrossRef Y. Doi, M. Yashiro, N. Yamada, R. Amano, G. Ohira, M. Komoto, S. Noda, S. Kashiwagi, Y. Kato, Y. Fuyuhiro, Significance of phospho-vascular endothelial growth factor receptor-2 expression in pancreatic cancer. Cancer Sci. 101, 1529–1535 (2010)PubMedCrossRef
23.
Zurück zum Zitat R.-F. Tang, S.-X. Wang, L. Peng, S.-X. Wang, M. Zhang, Z.-F. Li, Z.-M. Zhang, Y. Xiao, F.-R. Zhang, Expression of vascular endothelial growth factors A and C in human pancreatic cancer. World J. Gastroenterol. 12, 280 (2006)PubMedPubMedCentralCrossRef R.-F. Tang, S.-X. Wang, L. Peng, S.-X. Wang, M. Zhang, Z.-F. Li, Z.-M. Zhang, Y. Xiao, F.-R. Zhang, Expression of vascular endothelial growth factors A and C in human pancreatic cancer. World J. Gastroenterol. 12, 280 (2006)PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Z. Von Marschall, A. Scholz, S.A. Stacker, M.G. Achen, D.G. Jackson, F. Alves, M. Schirner, M. Haberey, K.-H. Thierauch, B. Wiedenmann, Vascular endothelial growth factor-D induces lymphangiogenesis and lymphatic metastasis in models of ductal pancreatic cancer. Int. J. Oncol. 27, 669–679 (2005) Z. Von Marschall, A. Scholz, S.A. Stacker, M.G. Achen, D.G. Jackson, F. Alves, M. Schirner, M. Haberey, K.-H. Thierauch, B. Wiedenmann, Vascular endothelial growth factor-D induces lymphangiogenesis and lymphatic metastasis in models of ductal pancreatic cancer. Int. J. Oncol. 27, 669–679 (2005)
25.
Zurück zum Zitat Y. Seo, H. Baba, T. Fukuda, M. Takashima, K. Sugimachi, High expression of vascular endothelial growth factor is associated with liver metastasis and a poor prognosis for patients with ductal pancreatic adenocarcinoma. Cancer 88, 2239–2245 (2000)PubMedCrossRef Y. Seo, H. Baba, T. Fukuda, M. Takashima, K. Sugimachi, High expression of vascular endothelial growth factor is associated with liver metastasis and a poor prognosis for patients with ductal pancreatic adenocarcinoma. Cancer 88, 2239–2245 (2000)PubMedCrossRef
26.
Zurück zum Zitat A. Stathis, F. Bertoni, BET proteins as targets for anticancer treatment. Cancer Discov. 8, 24–36 (2018)PubMedCrossRef A. Stathis, F. Bertoni, BET proteins as targets for anticancer treatment. Cancer Discov. 8, 24–36 (2018)PubMedCrossRef
27.
Zurück zum Zitat A. Ianevski, L. He, T. Aittokallio, J. Tang, SynergyFinder: a web application for analyzing drug combination dose–response matrix data. Bioinformatics 33, 2413–2415 (2017)PubMedPubMedCentralCrossRef A. Ianevski, L. He, T. Aittokallio, J. Tang, SynergyFinder: a web application for analyzing drug combination dose–response matrix data. Bioinformatics 33, 2413–2415 (2017)PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Q. Liu, X. Yin, L.R. Languino, D.C. Altieri, Evaluation of drug combination effect using a Bliss independence dose–response surface model. Stat. Biopharm. Res. 10, 112–122 (2018)PubMedPubMedCentralCrossRef Q. Liu, X. Yin, L.R. Languino, D.C. Altieri, Evaluation of drug combination effect using a Bliss independence dose–response surface model. Stat. Biopharm. Res. 10, 112–122 (2018)PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat M. Momeny, H. Yousefi, H. Eyvani, F. Moghaddaskho, A. Salehi, F. Esmaeili, Z. Alishahi, F. Barghi, S. Vaezijoze, S. Shamsaiegahkani, Blockade of nuclear factor-κB (NF-κB) pathway inhibits growth and induces apoptosis in chemoresistant ovarian carcinoma cells. Int. J. Biochem. Cell. Biol. 99, 1–9 (2018)CrossRef M. Momeny, H. Yousefi, H. Eyvani, F. Moghaddaskho, A. Salehi, F. Esmaeili, Z. Alishahi, F. Barghi, S. Vaezijoze, S. Shamsaiegahkani, Blockade of nuclear factor-κB (NF-κB) pathway inhibits growth and induces apoptosis in chemoresistant ovarian carcinoma cells. Int. J. Biochem. Cell. Biol. 99, 1–9 (2018)CrossRef
30.
Zurück zum Zitat M. Momeny, J.M. Saunus, F. Marturana, A.E.M. Reed, D. Black, G. Sala, S. Iacobelli, J.D. Holland, D. Yu, L. Da Silva, Heregulin-HER3-HER2 signaling promotes matrix metalloproteinase-dependent blood-brain-barrier transendothelial migration of human breast cancer cell lines. Oncotarget 6, 3932 (2015)PubMedPubMedCentralCrossRef M. Momeny, J.M. Saunus, F. Marturana, A.E.M. Reed, D. Black, G. Sala, S. Iacobelli, J.D. Holland, D. Yu, L. Da Silva, Heregulin-HER3-HER2 signaling promotes matrix metalloproteinase-dependent blood-brain-barrier transendothelial migration of human breast cancer cell lines. Oncotarget 6, 3932 (2015)PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat D.D. Von Hoff, D. Goldstein, M.F. Renschler, Albumin-bound paclitaxel plus gemcitabine in pancreatic cancer. N. Engl. J. Med. 370, 479 (2014) D.D. Von Hoff, D. Goldstein, M.F. Renschler, Albumin-bound paclitaxel plus gemcitabine in pancreatic cancer. N. Engl. J. Med. 370, 479 (2014)
33.
Zurück zum Zitat G. Kim, nab-Paclitaxel for the treatment of pancreatic cancer. Cancer Manag. Res. 9, 85 (2017) G. Kim, nab-Paclitaxel for the treatment of pancreatic cancer. Cancer Manag. Res. 9, 85 (2017)
34.
Zurück zum Zitat N.A. Franken, H.M. Rodermond, J. Stap, J. Haveman, C. Van Bree, Clonogenic assay of cells in vitro. Nat. Protoc. 1, 2315 (2006)PubMedCrossRef N.A. Franken, H.M. Rodermond, J. Stap, J. Haveman, C. Van Bree, Clonogenic assay of cells in vitro. Nat. Protoc. 1, 2315 (2006)PubMedCrossRef
35.
Zurück zum Zitat D. Murthy, K.S. Attri, P.K. Singh, Phosphoinositide 3-kinase signaling pathway in pancreatic ductal adenocarcinoma progression, pathogenesis, and therapeutics. Front. Physiol. 9, 335 (2018)PubMedPubMedCentralCrossRef D. Murthy, K.S. Attri, P.K. Singh, Phosphoinositide 3-kinase signaling pathway in pancreatic ductal adenocarcinoma progression, pathogenesis, and therapeutics. Front. Physiol. 9, 335 (2018)PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat C. Zhang, X. Sun, Y. Ren, Y. Lou, J. Zhou, M. Liu, D. Li, Validation of Polo-like kinase 1 as a therapeutic target in pancreatic cancer cells. Cancer Biol. Ther. 13, 1214–1220 (2012)PubMedPubMedCentralCrossRef C. Zhang, X. Sun, Y. Ren, Y. Lou, J. Zhou, M. Liu, D. Li, Validation of Polo-like kinase 1 as a therapeutic target in pancreatic cancer cells. Cancer Biol. Ther. 13, 1214–1220 (2012)PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat M. Hassan, H. Watari, A. AbuAlmaaty, Y. Ohba, N. Sakuragi, Apoptosis and molecular targeting therapy in cancer. Biomed. Res. Int. 2014, 150845 (2014) M. Hassan, H. Watari, A. AbuAlmaaty, Y. Ohba, N. Sakuragi, Apoptosis and molecular targeting therapy in cancer. Biomed. Res. Int. 2014, 150845 (2014)
39.
Zurück zum Zitat A.K. Nalla, B. Gorantla, C.S. Gondi, S.S. Lakka, J.S. Rao, Targeting MMP-9, uPAR, and cathepsin B inhibits invasion, migration and activates apoptosis in prostate cancer cells. Cancer Gene Ther. 17, 599 (2010)PubMedPubMedCentralCrossRef A.K. Nalla, B. Gorantla, C.S. Gondi, S.S. Lakka, J.S. Rao, Targeting MMP-9, uPAR, and cathepsin B inhibits invasion, migration and activates apoptosis in prostate cancer cells. Cancer Gene Ther. 17, 599 (2010)PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat J.-H. Hwang, S.H. Lee, K.H. Lee, K.Y. Lee, H. Kim, J.K. Ryu, Y.B. Yoon, Y.-T. Kim, Cathepsin B is a target of Hedgehog signaling in pancreatic cancer. Cancer Lett. 273, 266–272 (2009)PubMedCrossRef J.-H. Hwang, S.H. Lee, K.H. Lee, K.Y. Lee, H. Kim, J.K. Ryu, Y.B. Yoon, Y.-T. Kim, Cathepsin B is a target of Hedgehog signaling in pancreatic cancer. Cancer Lett. 273, 266–272 (2009)PubMedCrossRef
41.
Zurück zum Zitat V. Ellenrieder, S.F. Hendler, C. Ruhland, W. Boeck, G. Adler, T.M. Gress, TGF-β–induced invasiveness of pancreatic cancer cells is mediated by matrix metalloproteinase-2 and the urokinase plasminogen activator system. Int. J. Cancer. 93, 204–211 (2001)PubMedCrossRef V. Ellenrieder, S.F. Hendler, C. Ruhland, W. Boeck, G. Adler, T.M. Gress, TGF-β–induced invasiveness of pancreatic cancer cells is mediated by matrix metalloproteinase-2 and the urokinase plasminogen activator system. Int. J. Cancer. 93, 204–211 (2001)PubMedCrossRef
42.
Zurück zum Zitat T.W. Bauer, W. Liu, F. Fan, E.R. Camp, A. Yang, R.J. Somcio, C.D. Bucana, J. Callahan, G.C. Parry, D.B. Evans, Targeting of urokinase plasminogen activator receptor in human pancreatic carcinoma cells inhibits c-Met–and insulin-like growth factor-I receptor–mediated migration and invasion and orthotopic tumor growth in mice. Cancer Res. 65, 7775–7781 (2005)PubMedCrossRef T.W. Bauer, W. Liu, F. Fan, E.R. Camp, A. Yang, R.J. Somcio, C.D. Bucana, J. Callahan, G.C. Parry, D.B. Evans, Targeting of urokinase plasminogen activator receptor in human pancreatic carcinoma cells inhibits c-Met–and insulin-like growth factor-I receptor–mediated migration and invasion and orthotopic tumor growth in mice. Cancer Res. 65, 7775–7781 (2005)PubMedCrossRef
43.
Zurück zum Zitat D. Cantero, H. Friess, J. Deflorin, A. Zimmermann, M. Bründler, E. Riesle, M. Korc, M. Büchler, Enhanced expression of urokinase plasminogen activator and its receptor in pancreatic carcinoma. Br. J. Cancer 75, 388 (1997)PubMedPubMedCentralCrossRef D. Cantero, H. Friess, J. Deflorin, A. Zimmermann, M. Bründler, E. Riesle, M. Korc, M. Büchler, Enhanced expression of urokinase plasminogen activator and its receptor in pancreatic carcinoma. Br. J. Cancer 75, 388 (1997)PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat M. Bloomston, E.E. Zervos, A.S. Rosemurgy, Matrix metalloproteinases and their role in pancreatic cancer: a review of preclinical studies and clinical trials. Ann. Surg. Oncol. 9, 668–674 (2002)PubMedCrossRef M. Bloomston, E.E. Zervos, A.S. Rosemurgy, Matrix metalloproteinases and their role in pancreatic cancer: a review of preclinical studies and clinical trials. Ann. Surg. Oncol. 9, 668–674 (2002)PubMedCrossRef
45.
Zurück zum Zitat A. Gopinathan, G.M. DeNicola, K.K. Frese, N. Cook, F.A. Karreth, J. Mayerle, M.M. Lerch, T. Reinheckel, D.A. Tuveson, Cathepsin B promotes the progression of pancreatic ductal adenocarcinoma in mice. Gut 61, 877–884 (2012)PubMedCrossRef A. Gopinathan, G.M. DeNicola, K.K. Frese, N. Cook, F.A. Karreth, J. Mayerle, M.M. Lerch, T. Reinheckel, D.A. Tuveson, Cathepsin B promotes the progression of pancreatic ductal adenocarcinoma in mice. Gut 61, 877–884 (2012)PubMedCrossRef
46.
Zurück zum Zitat L. Dumartin, H.J. Whiteman, M.E. Weeks, D. Hariharan, B. Dmitrovic, C.A. Iacobuzio-Donahue, T.A. Brentnall, M.P. Bronner, R.M. Feakins, J.F. Timms, AGR2 is a novel surface antigen that promotes the dissemination of pancreatic cancer cells through regulation of cathepsins B and D. Cancer Res. 71, 7091–7102 (2011)PubMedPubMedCentralCrossRef L. Dumartin, H.J. Whiteman, M.E. Weeks, D. Hariharan, B. Dmitrovic, C.A. Iacobuzio-Donahue, T.A. Brentnall, M.P. Bronner, R.M. Feakins, J.F. Timms, AGR2 is a novel surface antigen that promotes the dissemination of pancreatic cancer cells through regulation of cathepsins B and D. Cancer Res. 71, 7091–7102 (2011)PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat T. Li, Y. Zhu, L. Han, W. Ren, H. Liu, C. Qin, VEGFR-1 activation-induced MMP-9-dependent invasion in hepatocellular carcinoma. Future Oncol. 11, 3143–3157 (2015)PubMedPubMedCentralCrossRef T. Li, Y. Zhu, L. Han, W. Ren, H. Liu, C. Qin, VEGFR-1 activation-induced MMP-9-dependent invasion in hepatocellular carcinoma. Future Oncol. 11, 3143–3157 (2015)PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat M. Momeny, F. Moghaddaskho, N.K. Gortany, H. Yousefi, Z. Sabourinejad, G. Zarrinrad, S. Mirshahvaladi, H. Eyvani, F. Barghi, L. Ahmadinia, Blockade of vascular endothelial growth factor receptors by tivozanib has potential anti-tumour effects on human glioblastoma cells. Sci. Rep. 7, 44075 (2017)PubMedPubMedCentralCrossRef M. Momeny, F. Moghaddaskho, N.K. Gortany, H. Yousefi, Z. Sabourinejad, G. Zarrinrad, S. Mirshahvaladi, H. Eyvani, F. Barghi, L. Ahmadinia, Blockade of vascular endothelial growth factor receptors by tivozanib has potential anti-tumour effects on human glioblastoma cells. Sci. Rep. 7, 44075 (2017)PubMedPubMedCentralCrossRef
49.
50.
Zurück zum Zitat P. Zhou, B. Li, F. Liu, M. Zhang, Q. Wang, Y. Liu, Y. Yao, D. Li, The epithelial to mesenchymal transition (EMT) and cancer stem cells: implication for treatment resistance in pancreatic cancer. Mol. Cancer. 16, 52 (2017)PubMedPubMedCentralCrossRef P. Zhou, B. Li, F. Liu, M. Zhang, Q. Wang, Y. Liu, Y. Yao, D. Li, The epithelial to mesenchymal transition (EMT) and cancer stem cells: implication for treatment resistance in pancreatic cancer. Mol. Cancer. 16, 52 (2017)PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat E.P. Balaban, P.B. Mangu, A.A. Khorana, M.A. Shah, S. Mukherjee, C.H. Crane, M.M. Javle, J.R. Eads, P. Allen, A.H. Ko, Locally advanced, unresectable pancreatic cancer: American Society of Clinical Oncology clinical practice guideline. J. Clin. Oncol. 34, 2654–2668 (2016)PubMedCrossRef E.P. Balaban, P.B. Mangu, A.A. Khorana, M.A. Shah, S. Mukherjee, C.H. Crane, M.M. Javle, J.R. Eads, P. Allen, A.H. Ko, Locally advanced, unresectable pancreatic cancer: American Society of Clinical Oncology clinical practice guideline. J. Clin. Oncol. 34, 2654–2668 (2016)PubMedCrossRef
52.
Zurück zum Zitat H.R. Cardenes, E.G. Chiorean, J. DeWitt, M. Schmidt, P. Loehrer, Locally advanced pancreatic cancer: current therapeutic approach. The Oncologist 11, 612–623 (2006)PubMedCrossRef H.R. Cardenes, E.G. Chiorean, J. DeWitt, M. Schmidt, P. Loehrer, Locally advanced pancreatic cancer: current therapeutic approach. The Oncologist 11, 612–623 (2006)PubMedCrossRef
53.
Zurück zum Zitat F. Wang, P. Kumar, The role of radiotherapy in management of pancreatic cancer. J. Gastrointest. Oncol. 2, 157 (2011)PubMedPubMedCentral F. Wang, P. Kumar, The role of radiotherapy in management of pancreatic cancer. J. Gastrointest. Oncol. 2, 157 (2011)PubMedPubMedCentral
54.
Zurück zum Zitat P. Seshacharyulu, M.J. Baine, J.J. Souchek, M. Menning, S. Kaur, Y. Yan, M.M. Ouellette, M. Jain, C. Lin, S.K. Batra, Biological determinants of radioresistance and their remediation in pancreatic cancer. Biochim. Biophys. Acta Rev. Cancer 1868, 69–92 (2017)CrossRef P. Seshacharyulu, M.J. Baine, J.J. Souchek, M. Menning, S. Kaur, Y. Yan, M.M. Ouellette, M. Jain, C. Lin, S.K. Batra, Biological determinants of radioresistance and their remediation in pancreatic cancer. Biochim. Biophys. Acta Rev. Cancer 1868, 69–92 (2017)CrossRef
55.
Zurück zum Zitat P. Wachsberger, R. Burd, A.P. Dicker, Tumor response to ionizing radiation combined with antiangiogenesis or vascular targeting agents: exploring mechanisms of interaction. Clin. Cancer Res. 9, 1957–1971 (2003)PubMed P. Wachsberger, R. Burd, A.P. Dicker, Tumor response to ionizing radiation combined with antiangiogenesis or vascular targeting agents: exploring mechanisms of interaction. Clin. Cancer Res. 9, 1957–1971 (2003)PubMed
56.
Zurück zum Zitat W. Zhao, L. Li, X. Zhu, L. Chen, X. Mo, K. Nie, Y. Lin, VEGF functions as a key modulator in the radioresistance formation of A549 cell lines via regulation of Notch1 expression. Int. J. Clin. Exp. Pathol. 10, 1627–1634 (2017) W. Zhao, L. Li, X. Zhu, L. Chen, X. Mo, K. Nie, Y. Lin, VEGF functions as a key modulator in the radioresistance formation of A549 cell lines via regulation of Notch1 expression. Int. J. Clin. Exp. Pathol. 10, 1627–1634 (2017)
57.
Zurück zum Zitat R. Mazeron, B. Anderson, S. Supiot, F. Paris, E. Deutsch, Current state of knowledge regarding the use of antiangiogenic agents with radiation therapy. Cancer Treat. Rev. 37, 476–486 (2011)PubMed R. Mazeron, B. Anderson, S. Supiot, F. Paris, E. Deutsch, Current state of knowledge regarding the use of antiangiogenic agents with radiation therapy. Cancer Treat. Rev. 37, 476–486 (2011)PubMed
58.
Zurück zum Zitat V. Longo, O. Brunetti, A. Gnoni, S. Cascinu, G. Gasparini, V. Lorusso, D. Ribatti, N. Silvestris, Angiogenesis in pancreatic ductal adenocarcinoma: a controversial issue. Oncotarget 7, 58649 (2016)PubMedPubMedCentralCrossRef V. Longo, O. Brunetti, A. Gnoni, S. Cascinu, G. Gasparini, V. Lorusso, D. Ribatti, N. Silvestris, Angiogenesis in pancreatic ductal adenocarcinoma: a controversial issue. Oncotarget 7, 58649 (2016)PubMedPubMedCentralCrossRef
59.
Zurück zum Zitat H. Kurahara, S. Takao, K. Maemura, H. Shinchi, S. Natsugoe, T. Aikou, Impact of vascular endothelial growth factor-C and-D expression in human pancreatic cancer: its relationship to lymph node metastasis. Clin. Cancer Res. 10, 8413–8420 (2004)PubMedCrossRef H. Kurahara, S. Takao, K. Maemura, H. Shinchi, S. Natsugoe, T. Aikou, Impact of vascular endothelial growth factor-C and-D expression in human pancreatic cancer: its relationship to lymph node metastasis. Clin. Cancer Res. 10, 8413–8420 (2004)PubMedCrossRef
60.
Zurück zum Zitat M.I. Costache, S. Iordache, C.A. Costache, E. Dragos, A. Dragos, A. Săftoiu, Molecular analysis of vascular endothelial growth factor (VEGF) receptors in EUS-guided samples obtained from patients with pancreatic adenocarcinoma. J. Gastrointestin. Liver Dis. 26, 51-57 (2017) M.I. Costache, S. Iordache, C.A. Costache, E. Dragos, A. Dragos, A. Săftoiu, Molecular analysis of vascular endothelial growth factor (VEGF) receptors in EUS-guided samples obtained from patients with pancreatic adenocarcinoma. J. Gastrointestin. Liver Dis. 26, 51-57 (2017)
61.
Zurück zum Zitat N. Ferrara, Vascular endothelial growth factor as a target for anticancer therapy. The Oncologist 9, 2–10 (2004)PubMedCrossRef N. Ferrara, Vascular endothelial growth factor as a target for anticancer therapy. The Oncologist 9, 2–10 (2004)PubMedCrossRef
62.
Zurück zum Zitat S. Dias, M. Choy, K. Alitalo, S. Rafii, Vascular endothelial growth factor (VEGF)–C signaling through FLT-4 (VEGFR-3) mediates leukemic cell proliferation, survival, and resistance to chemotherapy. Blood 99, 2179–2184 (2002)PubMedCrossRef S. Dias, M. Choy, K. Alitalo, S. Rafii, Vascular endothelial growth factor (VEGF)–C signaling through FLT-4 (VEGFR-3) mediates leukemic cell proliferation, survival, and resistance to chemotherapy. Blood 99, 2179–2184 (2002)PubMedCrossRef
63.
Zurück zum Zitat H. Zhu, F. Yun, X. Shi, D. Wang, VEGF-C inhibition reverses resistance of bladder cancer cells to cisplatin via upregulating maspin. Mol. Med. Rep. 12, 3163–3169 (2015)PubMedCrossRef H. Zhu, F. Yun, X. Shi, D. Wang, VEGF-C inhibition reverses resistance of bladder cancer cells to cisplatin via upregulating maspin. Mol. Med. Rep. 12, 3163–3169 (2015)PubMedCrossRef
64.
Zurück zum Zitat M. Momeny, Z. Sabourinejad, G. Zarrinrad, F. Moghaddaskho, H. Eyvani, H. Yousefi, S. Mirshahvaladi, E.M. Poursani, F. Barghi, A. Poursheikhani, Anti-tumour activity of tivozanib, a pan-inhibitor of VEGF receptors, in therapy-resistant ovarian carcinoma cells. Sci. Rep. 7, 45954 (2017)PubMedPubMedCentralCrossRef M. Momeny, Z. Sabourinejad, G. Zarrinrad, F. Moghaddaskho, H. Eyvani, H. Yousefi, S. Mirshahvaladi, E.M. Poursani, F. Barghi, A. Poursheikhani, Anti-tumour activity of tivozanib, a pan-inhibitor of VEGF receptors, in therapy-resistant ovarian carcinoma cells. Sci. Rep. 7, 45954 (2017)PubMedPubMedCentralCrossRef
66.
Zurück zum Zitat Q. Wu, L. Miele, F.H. Sarkar, Z. Wang, The role of EMT in pancreatic cancer progression. Pancreat Disord Ther. 2, e121 (2012) Q. Wu, L. Miele, F.H. Sarkar, Z. Wang, The role of EMT in pancreatic cancer progression. Pancreat Disord Ther. 2, e121 (2012)
67.
Zurück zum Zitat B. Hotz, M. Arndt, S. Dullat, S. Bhargava, H.-J. Buhr, H.G. Hotz, Epithelial to mesenchymal transition: expression of the regulators snail, slug, and twist in pancreatic cancer. Clin. Cancer Res. 13, 4769–4776 (2007)PubMedCrossRef B. Hotz, M. Arndt, S. Dullat, S. Bhargava, H.-J. Buhr, H.G. Hotz, Epithelial to mesenchymal transition: expression of the regulators snail, slug, and twist in pancreatic cancer. Clin. Cancer Res. 13, 4769–4776 (2007)PubMedCrossRef
68.
Zurück zum Zitat J.M. Ebos, C.R. Lee, W. Cruz-Munoz, G.A. Bjarnason, J.G. Christensen, R.S. Kerbel, Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 15, 232–239 (2009)PubMedPubMedCentralCrossRef J.M. Ebos, C.R. Lee, W. Cruz-Munoz, G.A. Bjarnason, J.G. Christensen, R.S. Kerbel, Accelerated metastasis after short-term treatment with a potent inhibitor of tumor angiogenesis. Cancer Cell 15, 232–239 (2009)PubMedPubMedCentralCrossRef
69.
Zurück zum Zitat M. Kim, K. Jang, P. Miller, M. Picon-Ruiz, T. Yeasky, D. El-Ashry, J. Slingerland, VEGFA links self-renewal and metastasis by inducing Sox2 to repress miR-452, driving Slug. Oncogene 36, 5199 (2017)PubMedPubMedCentralCrossRef M. Kim, K. Jang, P. Miller, M. Picon-Ruiz, T. Yeasky, D. El-Ashry, J. Slingerland, VEGFA links self-renewal and metastasis by inducing Sox2 to repress miR-452, driving Slug. Oncogene 36, 5199 (2017)PubMedPubMedCentralCrossRef
70.
Zurück zum Zitat L. Marquez-Exposito, C. Lavoz, R.R. Rodrigues-Diez, S. Rayego-Mateos, M. Orejudo, E. Cantero-Navarro, A. Ortiz, J. Egido, R. Selgas, S. Mezzano, Gremlin regulates tubular epithelial to mesenchymal transition via VEGFR2: Potential role in renal fibrosis. Front. Pharmacol. 9, 1195 (2018)PubMedPubMedCentralCrossRef L. Marquez-Exposito, C. Lavoz, R.R. Rodrigues-Diez, S. Rayego-Mateos, M. Orejudo, E. Cantero-Navarro, A. Ortiz, J. Egido, R. Selgas, S. Mezzano, Gremlin regulates tubular epithelial to mesenchymal transition via VEGFR2: Potential role in renal fibrosis. Front. Pharmacol. 9, 1195 (2018)PubMedPubMedCentralCrossRef
71.
Zurück zum Zitat Y.-W. Yeh, C.-C. Cheng, S.-T. Yang, C.-F. Tseng, T.-Y. Chang, S.-Y. Tsai, E. Fu, C.-P. Chiang, L.-C. Liao, P.-W. Tsai, Targeting the VEGF-C/VEGFR3 axis suppresses Slug-mediated cancer metastasis and stemness via inhibition of KRAS/YAP1 signaling. Oncotarget 8, 5603 (2017)PubMed Y.-W. Yeh, C.-C. Cheng, S.-T. Yang, C.-F. Tseng, T.-Y. Chang, S.-Y. Tsai, E. Fu, C.-P. Chiang, L.-C. Liao, P.-W. Tsai, Targeting the VEGF-C/VEGFR3 axis suppresses Slug-mediated cancer metastasis and stemness via inhibition of KRAS/YAP1 signaling. Oncotarget 8, 5603 (2017)PubMed
74.
Zurück zum Zitat N. Samm, K. Werner, F. Rückert, H.D. Saeger, R. Grützmann, C. Pilarsky, The role of apoptosis in the pathology of pancreatic cancer. Cancers (Basel) 3, 1–16 (2010)PubMedPubMedCentralCrossRef N. Samm, K. Werner, F. Rückert, H.D. Saeger, R. Grützmann, C. Pilarsky, The role of apoptosis in the pathology of pancreatic cancer. Cancers (Basel) 3, 1–16 (2010)PubMedPubMedCentralCrossRef
75.
Zurück zum Zitat H. Friess, Z. Lu, A. Andrén-Sandberg, P. Berberat, A. Zimmermann, G. Adler, R. Schmid, M.W. Büchler, Moderate activation of the apoptosis inhibitor bcl-xL worsens the prognosis in pancreatic cancer. Ann. Surg. 228, 780 (1998)PubMedPubMedCentralCrossRef H. Friess, Z. Lu, A. Andrén-Sandberg, P. Berberat, A. Zimmermann, G. Adler, R. Schmid, M.W. Büchler, Moderate activation of the apoptosis inhibitor bcl-xL worsens the prognosis in pancreatic cancer. Ann. Surg. 228, 780 (1998)PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat S. Li, J. Sun, J. Yang, L. Zhang, L. Wang, X. Wang, Z. Guo, XIAP expression is associated with pancreatic carcinoma outcome. Mol. Clin. Oncol. 1, 305–308 (2013)PubMedPubMedCentralCrossRef S. Li, J. Sun, J. Yang, L. Zhang, L. Wang, X. Wang, Z. Guo, XIAP expression is associated with pancreatic carcinoma outcome. Mol. Clin. Oncol. 1, 305–308 (2013)PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Z. Chen, V. Sangwan, S. Banerjee, T. Mackenzie, V. Dudeja, X. Li, H. Wang, S.M. Vickers, A.K. Saluja, miR-204 mediated loss of Myeloid cell leukemia-1 results in pancreatic cancer cell death. Mol. Cancer 12, 105 (2013)PubMedPubMedCentralCrossRef Z. Chen, V. Sangwan, S. Banerjee, T. Mackenzie, V. Dudeja, X. Li, H. Wang, S.M. Vickers, A.K. Saluja, miR-204 mediated loss of Myeloid cell leukemia-1 results in pancreatic cancer cell death. Mol. Cancer 12, 105 (2013)PubMedPubMedCentralCrossRef
78.
Zurück zum Zitat N. Samm, K. Werner, F. Rückert, H.D. Saeger, R. Grützmann, C. Pilarsky, The role of apoptosis in the pathology of pancreatic cancer. Cancers (Basel) 3, 1–16 (2010)PubMedPubMedCentralCrossRef N. Samm, K. Werner, F. Rückert, H.D. Saeger, R. Grützmann, C. Pilarsky, The role of apoptosis in the pathology of pancreatic cancer. Cancers (Basel) 3, 1–16 (2010)PubMedPubMedCentralCrossRef
79.
Zurück zum Zitat S.-H. Wei, K. Dong, F. Lin, X. Wang, B. Li, J.-j. Shen, Q. Zhang, R. Wang, H.-Z. Zhang, Inducing apoptosis and enhancing chemosensitivity to gemcitabine via RNA interference targeting Mcl-1 gene in pancreatic carcinoma cell. Cancer Chemother. Pharmacol. 62, 1055–1064 (2008)PubMedCrossRef S.-H. Wei, K. Dong, F. Lin, X. Wang, B. Li, J.-j. Shen, Q. Zhang, R. Wang, H.-Z. Zhang, Inducing apoptosis and enhancing chemosensitivity to gemcitabine via RNA interference targeting Mcl-1 gene in pancreatic carcinoma cell. Cancer Chemother. Pharmacol. 62, 1055–1064 (2008)PubMedCrossRef
80.
Zurück zum Zitat Q. Guo, Y. Chen, B. Zhang, M. Kang, Q. Xie, Y. Wu, Potentiation of the effect of gemcitabine by emodin in pancreatic cancer is associated with survivin inhibition. Biochem. Pharmacol. 77, 1674–1683 (2009)PubMedCrossRef Q. Guo, Y. Chen, B. Zhang, M. Kang, Q. Xie, Y. Wu, Potentiation of the effect of gemcitabine by emodin in pancreatic cancer is associated with survivin inhibition. Biochem. Pharmacol. 77, 1674–1683 (2009)PubMedCrossRef
81.
Zurück zum Zitat J. Bai, J. Sui, A. Demirjian, C.M. Vollmer, W. Marasco, M.P. Callery, Predominant Bcl-XL knockdown disables antiapoptotic mechanisms: Tumor necrosis factor–related apoptosis-inducing ligand–based triple chemotherapy overcomes chemoresistance in pancreatic cancer cells in vitro. Cancer Res. 65, 2344–2352 (2005)PubMedCrossRef J. Bai, J. Sui, A. Demirjian, C.M. Vollmer, W. Marasco, M.P. Callery, Predominant Bcl-XL knockdown disables antiapoptotic mechanisms: Tumor necrosis factor–related apoptosis-inducing ligand–based triple chemotherapy overcomes chemoresistance in pancreatic cancer cells in vitro. Cancer Res. 65, 2344–2352 (2005)PubMedCrossRef
82.
Zurück zum Zitat H. Takahashi, M.C. Chen, H. Pham, Y. Matsuo, H. Ishiguro, H.A. Reber, H. Takeyama, O.J. Hines, G. Eibl, Simultaneous knock-down of Bcl-xL and Mcl-1 induces apoptosis through Bax activation in pancreatic cancer cells. Biochim. Biophys. Acta 1833, 2980–2987 (2013)CrossRef H. Takahashi, M.C. Chen, H. Pham, Y. Matsuo, H. Ishiguro, H.A. Reber, H. Takeyama, O.J. Hines, G. Eibl, Simultaneous knock-down of Bcl-xL and Mcl-1 induces apoptosis through Bax activation in pancreatic cancer cells. Biochim. Biophys. Acta 1833, 2980–2987 (2013)CrossRef
83.
Zurück zum Zitat E.J. Van Limbergen, P. Zabrocki, M. Porcu, E. Hauben, J. Cools, S. Nuyts, FLT1 kinase is a mediator of radioresistance and survival in head and neck squamous cell carcinoma. Acta Ocologica 53, 637–645 (2014) E.J. Van Limbergen, P. Zabrocki, M. Porcu, E. Hauben, J. Cools, S. Nuyts, FLT1 kinase is a mediator of radioresistance and survival in head and neck squamous cell carcinoma. Acta Ocologica 53, 637–645 (2014)
84.
Zurück zum Zitat P. Knizetova, J. Ehrmann, A. Hlobilkova, I. Vancova, O. Kalita, Z. Kolar, J. Bartek, Autocrine regulation of glioblastoma cell-cycle progression, viability and radioresistance through the VEGF-VEGFR2 (KDR) interplay. Cell Cycle 7, 2553–2561 (2008)PubMedCrossRef P. Knizetova, J. Ehrmann, A. Hlobilkova, I. Vancova, O. Kalita, Z. Kolar, J. Bartek, Autocrine regulation of glioblastoma cell-cycle progression, viability and radioresistance through the VEGF-VEGFR2 (KDR) interplay. Cell Cycle 7, 2553–2561 (2008)PubMedCrossRef
85.
Zurück zum Zitat S. Huang, H. Ma, Effect of radiotherapy on angiogenesis of human pancreatic cancer transplanted tumor in nude mice. The Chinese-German Journal of Clinical Oncology 11, 635–637 (2012)CrossRef S. Huang, H. Ma, Effect of radiotherapy on angiogenesis of human pancreatic cancer transplanted tumor in nude mice. The Chinese-German Journal of Clinical Oncology 11, 635–637 (2012)CrossRef
86.
Zurück zum Zitat K. Yoshida, S. Suzuki, J. Sakata, F. Utsumi, K. Niimi, N. Yoshikawa, K. Nishino, K. Shibata, F. Kikkawa, H. Kajiyama, The upregulated expression of vascular endothelial growth factor in surgically treated patients with recurrent/radioresistant cervical cancer of the uterus. Oncol. Lett. 16, 515–521 (2018)PubMedPubMedCentral K. Yoshida, S. Suzuki, J. Sakata, F. Utsumi, K. Niimi, N. Yoshikawa, K. Nishino, K. Shibata, F. Kikkawa, H. Kajiyama, The upregulated expression of vascular endothelial growth factor in surgically treated patients with recurrent/radioresistant cervical cancer of the uterus. Oncol. Lett. 16, 515–521 (2018)PubMedPubMedCentral
87.
Zurück zum Zitat Y.-H. Chen, S.-L. Pan, J.-C. Wang, S.-H. Kuo, J.C.-H. Cheng, C.-M. Teng, Radiation-induced VEGF-C expression and endothelial cell proliferation in lung cancer. Strahlenther. Onkol. 190, 1154–1162 (2014)PubMedPubMedCentralCrossRef Y.-H. Chen, S.-L. Pan, J.-C. Wang, S.-H. Kuo, J.C.-H. Cheng, C.-M. Teng, Radiation-induced VEGF-C expression and endothelial cell proliferation in lung cancer. Strahlenther. Onkol. 190, 1154–1162 (2014)PubMedPubMedCentralCrossRef
88.
Zurück zum Zitat H.-W. Hsu, N.R. Wall, C.-T. Hsueh, S. Kim, R.L. Ferris, C.-S. Chen, S. Mirshahidi, Combination antiangiogenic therapy and radiation in head and neck cancers. Oral Oncol. 50, 19–26 (2014)PubMedCrossRef H.-W. Hsu, N.R. Wall, C.-T. Hsueh, S. Kim, R.L. Ferris, C.-S. Chen, S. Mirshahidi, Combination antiangiogenic therapy and radiation in head and neck cancers. Oral Oncol. 50, 19–26 (2014)PubMedCrossRef
89.
Zurück zum Zitat C.H. Crane, K. Winter, W.F. Regine, H. Safran, T.A. Rich, W. Curran, R.A. Wolff, C.G. Willett, Phase II study of bevacizumab with concurrent capecitabine and radiation followed by maintenance gemcitabine and bevacizumab for locally advanced pancreatic cancer: Radiation Therapy Oncology Group RTOG 0411. J. Clin. Oncol. 27, 4096 (2009)PubMedPubMedCentralCrossRef C.H. Crane, K. Winter, W.F. Regine, H. Safran, T.A. Rich, W. Curran, R.A. Wolff, C.G. Willett, Phase II study of bevacizumab with concurrent capecitabine and radiation followed by maintenance gemcitabine and bevacizumab for locally advanced pancreatic cancer: Radiation Therapy Oncology Group RTOG 0411. J. Clin. Oncol. 27, 4096 (2009)PubMedPubMedCentralCrossRef
90.
Zurück zum Zitat A. Affolter, G. Samosny, A.S. Heimes, J. Schneider, W. Weichert, A. Stenzinger, K. Sommer, A. Jensen, A. Mayer, W. Brenner, Multikinase inhibitors sorafenib and sunitinib as radiosensitizers in head and neck cancer cell lines. Head & Neck 39, 623–632 (2017) A. Affolter, G. Samosny, A.S. Heimes, J. Schneider, W. Weichert, A. Stenzinger, K. Sommer, A. Jensen, A. Mayer, W. Brenner, Multikinase inhibitors sorafenib and sunitinib as radiosensitizers in head and neck cancer cell lines. Head & Neck 39, 623–632 (2017)
91.
Zurück zum Zitat D. Wei, Q. Zhang, J.S. Schreiber, L.A. Parsels, F.A. Abulwerdi, T. Kausar, T.S. Lawrence, Y. Sun, Z. Nikolovska-Coleska, M.A. Morgan, Targeting mcl-1 for radiosensitization of pancreatic cancers. Transl. Oncol. 8, 47–54 (2015)PubMedPubMedCentralCrossRef D. Wei, Q. Zhang, J.S. Schreiber, L.A. Parsels, F.A. Abulwerdi, T. Kausar, T.S. Lawrence, Y. Sun, Z. Nikolovska-Coleska, M.A. Morgan, Targeting mcl-1 for radiosensitization of pancreatic cancers. Transl. Oncol. 8, 47–54 (2015)PubMedPubMedCentralCrossRef
92.
Zurück zum Zitat K. Kami, R. Doi, M. Koizumi, E. Toyoda, T. Mori, D. Ito, Y. Kawaguchi, K. Fujimoto, M. Wada, S.-I. Miyatake, Downregulation of survivin by siRNA diminishes radioresistance of pancreatic cancer cells. Surgery 138, 299–305 (2005)PubMedCrossRef K. Kami, R. Doi, M. Koizumi, E. Toyoda, T. Mori, D. Ito, Y. Kawaguchi, K. Fujimoto, M. Wada, S.-I. Miyatake, Downregulation of survivin by siRNA diminishes radioresistance of pancreatic cancer cells. Surgery 138, 299–305 (2005)PubMedCrossRef
93.
Zurück zum Zitat S.R. Wedge, J. Kendrew, L.F. Hennequin, P.J. Valentine, S.T. Barry, S.R. Brave, N.R. Smith, N.H. James, M. Dukes, J.O. Curwen, AZD2171: a highly potent, orally bioavailable, vascular endothelial growth factor receptor-2 tyrosine kinase inhibitor for the treatment of cancer. Cancer Res. 65, 4389–4400 (2005)PubMedCrossRef S.R. Wedge, J. Kendrew, L.F. Hennequin, P.J. Valentine, S.T. Barry, S.R. Brave, N.R. Smith, N.H. James, M. Dukes, J.O. Curwen, AZD2171: a highly potent, orally bioavailable, vascular endothelial growth factor receptor-2 tyrosine kinase inhibitor for the treatment of cancer. Cancer Res. 65, 4389–4400 (2005)PubMedCrossRef
94.
Zurück zum Zitat J. Kendrew, R. Odedra, A. Logié, P.J. Taylor, S. Pearsall, D.J. Ogilvie, S.R. Wedge, J.M. Jürgensmeier, Anti-tumour and anti-vascular effects of cediranib (AZD2171) alone and in combination with other anti-tumour therapies. Cancer Chemother. Pharmacol. 71, 1021–1032 (2013)PubMedCrossRef J. Kendrew, R. Odedra, A. Logié, P.J. Taylor, S. Pearsall, D.J. Ogilvie, S.R. Wedge, J.M. Jürgensmeier, Anti-tumour and anti-vascular effects of cediranib (AZD2171) alone and in combination with other anti-tumour therapies. Cancer Chemother. Pharmacol. 71, 1021–1032 (2013)PubMedCrossRef
95.
Zurück zum Zitat I. Ruscito, M.L. Gasparri, C. Marchetti, C. De Medici, C. Bracchi, I. Palaia, S. Imboden, M.D. Mueller, A. Papadia, L. Muzii, Cediranib in ovarian cancer: state of the art and future perspectives. Tumor Biol. 37, 2833–2839 (2016)PubMedCrossRef I. Ruscito, M.L. Gasparri, C. Marchetti, C. De Medici, C. Bracchi, I. Palaia, S. Imboden, M.D. Mueller, A. Papadia, L. Muzii, Cediranib in ovarian cancer: state of the art and future perspectives. Tumor Biol. 37, 2833–2839 (2016)PubMedCrossRef
96.
Zurück zum Zitat R.P. Symonds, C. Gourley, S. Davidson, K. Carty, E. McCartney, D. Rai, S. Banerjee, D. Jackson, R. Lord, M. McCormack, Cediranib combined with carboplatin and paclitaxel in patients with metastatic or recurrent cervical cancer (CIRCCa): a randomised, double-blind, placebo-controlled phase 2 trial. Lancet Oncol. 16, 1515–1524 (2015)PubMedPubMedCentralCrossRef R.P. Symonds, C. Gourley, S. Davidson, K. Carty, E. McCartney, D. Rai, S. Banerjee, D. Jackson, R. Lord, M. McCormack, Cediranib combined with carboplatin and paclitaxel in patients with metastatic or recurrent cervical cancer (CIRCCa): a randomised, double-blind, placebo-controlled phase 2 trial. Lancet Oncol. 16, 1515–1524 (2015)PubMedPubMedCentralCrossRef
97.
Zurück zum Zitat W.L. Dahut, R.A. Madan, J.J. Karakunnel, D. Adelberg, J.L. Gulley, I.B. Turkbey, C.H. Chau, S.D. Spencer, M. Mulquin, J. Wright, Phase II clinical trial of cediranib in patients with metastatic castration-resistant prostate cancer. BJU Int. 111, 1269–1280 (2013)PubMedPubMedCentralCrossRef W.L. Dahut, R.A. Madan, J.J. Karakunnel, D. Adelberg, J.L. Gulley, I.B. Turkbey, C.H. Chau, S.D. Spencer, M. Mulquin, J. Wright, Phase II clinical trial of cediranib in patients with metastatic castration-resistant prostate cancer. BJU Int. 111, 1269–1280 (2013)PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat S.S. Sridhar, M.J. Mackenzie, S.J. Hotte, S.D. Mukherjee, I.F. Tannock, N. Murray, C. Kollmannsberger, M.A. Haider, E.X. Chen, R. Halford, A phase II study of cediranib (AZD 2171) in treatment naive patients with progressive unresectable recurrent or metastatic renal cell carcinoma. A trial of the PMH phase 2 consortium. Invest. New Drugs 31, 1008–1015 (2013)PubMedCrossRef S.S. Sridhar, M.J. Mackenzie, S.J. Hotte, S.D. Mukherjee, I.F. Tannock, N. Murray, C. Kollmannsberger, M.A. Haider, E.X. Chen, R. Halford, A phase II study of cediranib (AZD 2171) in treatment naive patients with progressive unresectable recurrent or metastatic renal cell carcinoma. A trial of the PMH phase 2 consortium. Invest. New Drugs 31, 1008–1015 (2013)PubMedCrossRef
Metadaten
Titel
Anti-tumor activity of cediranib, a pan-vascular endothelial growth factor receptor inhibitor, in pancreatic ductal adenocarcinoma cells
verfasst von
Majid Momeny
Zivar Alishahi
Haniyeh Eyvani
Fatemeh Esmaeili
Azam Zaghal
Parisa Ghaffari
Javad Tavakkoly-Bazzaz
Kamran Alimoghaddam
Ardeshir Ghavamzadeh
Seyed H. Ghaffari
Publikationsdatum
06.09.2019
Verlag
Springer Netherlands
Erschienen in
Cellular Oncology / Ausgabe 1/2020
Print ISSN: 2211-3428
Elektronische ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-019-00473-9

Weitere Artikel der Ausgabe 1/2020

Cellular Oncology 1/2020 Zur Ausgabe

Neu im Fachgebiet Pathologie

Assistierter Suizid durch Infusion von Thiopental

Thiopental Originalie

Als Folge des Urteils des Bundesverfassungsgerichts zur Sterbehilfe im Jahr 2020 wurde in den Jahren 2021–2023 eine Reihe (n = 23) von assistierten Suiziden im Landesinstitut für gerichtliche und soziale Medizin Berlin mit jeweils identischen …

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 …