Skip to main content

Advertisement

Log in

Antiangiogenic therapy with Nintedanib affects hypoxia, angiogenesis and apoptosis in the ventral prostate of TRAMP animals

  • Regular Article
  • Published:
Cell and Tissue Research Aims and scope Submit manuscript

Abstract

The antiangiogenic therapy for prostate cancer with Nintedanib, a potent inhibitor of important growth factor receptors, has been proven to delay tumor progression and arrest tumor growth; thus, the aim herein is to evaluate Nintedanib effects on tumor cells, besides angiogenesis and apoptosis processes, metalloproteinases and hypoxia factor in an animal model. Nintedanib promoted growth inhibition and cell death in a dose-dependent manner, showing no tumor selectivity. Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) were treated with Nintedanib (10 mg/kg/day) in different stages of tumor development and the ventral prostate was examined for protein levels by means of immunohistochemistry and Western blotting and apoptosis evaluation. In vitro antiproliferative activity of Nintedanib was also assessed in nine human tumor cell lines. Early Nintedanib treatment has shown decreased levels of FGF-2, VEGFR-1, MMP-9 and HIF-1α and a significantly increased apoptosis of epithelial cells. Furthermore, late Nintedanib treatment decreased FGF-2, VEGFR-1 and FGFR-3 levels. Importantly, even after treatment discontinuation, treated animals displayed a significant decrease in VEGFR-1 as well as MMP-9. Although Nintedanib treatment in late stages of tumor growth has shown some good results, it is noteworthy that the drug presents the best tissue response when administered in the early stages of disease development. Nintedanib treatment has shown to be a promising approach for prostate cancer therapy, especially in the early stages of the disease, interfering in different carcinogenesis progression pathways.

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

  • Alves LF, da Silva RF, Cagnon VHA (2018) Nintedanib effects on delaying cancer progression and decreasing COX-2 and IL-17 in the prostate anterior lobe in TRAMP mice. Tissue Cell 50:96–103

    CAS  PubMed  Google Scholar 

  • Awasthi N, Schwarz RE (2015) Profile of nintedanib in the treatment of solid tumors: the evidence to date. Onco Targets Ther 8:3691–3701

    CAS  PubMed  PubMed Central  Google Scholar 

  • Awasthi N, Hinz S, Brekken RA, Schwarz MA, Schwarz RE (2015) Nintedanib, a triple angiokinase inhibitor, enhances cytotoxic therapy response in pancreatic cancer. Cancer Lett 358:59–66

    CAS  PubMed  Google Scholar 

  • Boget S, Cereser C, Parvaz P, Leriche A, Revol A (2001) Fibroblast growth factor receptor 1 (FGFR1) is over-expressed in benign prostatic hyperplasia whereas FGFR2-IIIc and FGFR3 are not. Eur J Endocrinol 145:303–310

    CAS  PubMed  Google Scholar 

  • Castellano G, Malaponte G, Mazzarino MC, Figini M, Marchese F, Gangemi P, Travali S, Stivala F, Canevari S, Libra M (2008) Activation of the osteopontin/matrix metalloproteinase-9 pathway correlates with prostate cancer progression. Clin Cancer Res 14:7470–7480

    CAS  PubMed  Google Scholar 

  • da Silva RF, Nogueira-Pangrazi E, Kido LA, Montico F, Arana S, Kumar D, Raina K, Agarwal R, Cagnon VHA (2017) Nintedanib antiangiogenic inhibitor effectiveness in delaying adenocarcinoma progression in Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP). J Biomed Sci 24:31

    PubMed  PubMed Central  Google Scholar 

  • da Silva RF, Dhar D, Raina K, Kumar D, Kant R, Cagnon VHA, Agarwal C, Agarwal R (2018) Nintedanib inhibits growth of human prostate carcinoma cells by modulating both cell cycle and angiogenesis regulators. Sci Rep 8:9540

    PubMed  PubMed Central  Google Scholar 

  • Damasceno AA, Carvalho CP, Santos EM, Botelho FV, Araujo FA, Deconte SR, Tomiosso TC, Balbi AP, Zanon RG, Taboga SR, Goes RM, Ribeiro DL (2014) Effects of maternal diabetes on male offspring: high cell proliferation and increased activity of MMP-2 in the ventral prostate. Cell Tissue Res 358:257–269

    CAS  PubMed  Google Scholar 

  • Daniyal M, Siddiqui ZA, Akram M, Asif HM, Sultana S, Khan A (2014) Epidemiology, etiology, diagnosis and treatment of prostate cancer. Asian Pac J Cancer Prev 15:9575–9578

    PubMed  Google Scholar 

  • Doll JA, Reiher FK, Crawford SE, Pins MR, Campbell SC, Bouck NP (2001) Thrombospondin-1, vascular endothelial growth factor and fibroblast growth factor-2 are key functional regulators of angiogenesis in the prostate. Prostate 49:293–305

    CAS  PubMed  Google Scholar 

  • Feng S, Shao L, Yu W, Gavine P, Ittmann M (2012) Targeting fibroblast growth factor receptor signaling inhibits prostate cancer progression. Clin Cancer Res 18:3880–3888

    CAS  PubMed  PubMed Central  Google Scholar 

  • Feng S, Shao L, Castro P, Coleman I, Nelson PS, Smith PD, Davies BR, Ittmann M (2017) Combination treatment of prostate cancer with FGF receptor and AKT kinase inhibitors. Oncotarget 8:6179–6192

    PubMed  Google Scholar 

  • Ferrara N, Gerber HP, LeCouter J (2003) The biology of VEGF and its receptors. Nat Med 9:669–676

    CAS  PubMed  Google Scholar 

  • Giri D, Ropiquet F, Ittmann M (1999) Alterations in expression of basic fibroblast growth factor (FGF) 2 and its receptor FGFR-1 in human prostate cancer. Clin Cancer Res 5:1063–1071

    CAS  PubMed  Google Scholar 

  • Goncalves BF, de Campos SG, Zanetoni C, Scarano WR, Falleiros LR Jr, Amorim RL, Goes RM, Taboga SR (2013) A new proposed rodent model of chemically induced prostate carcinogenesis: distinct time-course prostate cancer progression in the dorsolateral and ventral lobes. Prostate 73:1202–1213

    CAS  PubMed  Google Scholar 

  • Gowardhan B, Douglas DA, Mathers ME, McKie AB, McCracken SR, Robson CN, Leung HY (2005) Evaluation of the fibroblast growth factor system as a potential target for therapy in human prostate cancer. Br J Cancer 92:320–327

    CAS  PubMed  PubMed Central  Google Scholar 

  • Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144:646–674

    CAS  Google Scholar 

  • Hernandez S, de Muga S, Agell L, Juanpere N, Esgueva R, Lorente JA, Mojal S, Serrano S, Lloreta J (2009) FGFR3 mutations in prostate cancer: association with low-grade tumors. Mod Pathol 22:848–856

    CAS  PubMed  Google Scholar 

  • Hilberg F, Roth GJ, Krssak M, Kautschitsch S, Sommergruber W, Tontsch-Grunt U, Garin-Chesa P, Bader G, Zoephel A, Quant J, Heckel A, Rettig WJ (2008) BIBF 1120: triple angiokinase inhibitor with sustained receptor blockade and good antitumor efficacy. Cancer Res 68:4774–4782

    CAS  PubMed  Google Scholar 

  • Huss WJ, Hanrahan CF, Barrios RJ, Simons JW, Greenberg NM (2001) Angiogenesis and prostate cancer: identification of a molecular progression switch. Cancer Res 61:2736–2743

    CAS  PubMed  Google Scholar 

  • Huss WJ, Barrios RJ, Foster BA, Greenberg NM (2003) Differential expression of specific FGF ligand and receptor isoforms during angiogenesis associated with prostate cancer progression. Prostate 54:8–16

    PubMed  Google Scholar 

  • Kudo K, Arao T, Tanaka K, Nagai T, Furuta K, Sakai K, Kaneda H, Matsumoto K, Tamura D, Aomatsu K, De Velasco MA, Fujita Y, Saijo N, Kudo M, Nishio K (2011) Antitumor activity of BIBF 1120, a triple angiokinase inhibitor and use of VEGFR2+pTyr+ peripheral blood leukocytes as a pharmacodynamic biomarker in vivo. Clin Cancer Res 17:1373–1381

    CAS  PubMed  Google Scholar 

  • Labi V, Erlacher M (2015) How cell death shapes cancer. Cell Death Dis 6:e1675

    CAS  PubMed  PubMed Central  Google Scholar 

  • Lo Iacono M, Buttigliero C, Monica V, Bollito E, Garrou D, Cappia S, Rapa I, Vignani F, Bertaglia V, Fiori C, Papotti M, Volante M, Scagliotti GV, Porpiglia F, Tucci M (2016) Retrospective study testing next generation sequencing of selected cancer-associated genes in resected prostate cancer. Oncotarget 7:14394–14404

    PubMed  PubMed Central  Google Scholar 

  • Lowe SW, Lin AW (2000) Apoptosis in cancer. Carcinogenesis 21:485–495

    CAS  PubMed  Google Scholar 

  • McCormack PL (2015) Nintedanib: first global approval. Drugs 75:129–139

    CAS  PubMed  Google Scholar 

  • Mollereau B, Perez-Garijo A, Bergmann A, Miura M, Gerlitz O, Ryoo HD, Steller H, Morata G (2013) Compensatory proliferation and apoptosis-induced proliferation: a need for clarification. Cell Death Differ 20:181

    CAS  PubMed  Google Scholar 

  • Monks A, Scudiero D, Skehan P, Shoemaker R, Paull K, Vistica D, Hose C, Langley J, Cronise P, Vaigro-Wolff A et al (1991) Feasibility of a high-flux anticancer drug screen using a diverse panel of cultured human tumor cell lines. J Natl Cancer Inst 83:757–766

    CAS  PubMed  Google Scholar 

  • Montico F, Kido LA, Hetzl AC, Lorencini RM, Candido EM, Cagnon VH (2014) Antiangiogenic therapy effects on age-associated matrix metalloproteinase-9 (MMP-9) and insulin-like growth factor receptor-1 (IGFR-1) responses: a comparative study of prostate disorders in aged and TRAMP mice. Histochem Cell Biol 142:269–284

    CAS  PubMed  Google Scholar 

  • Mukherji D, Temraz S, Wehbe D, Shamseddine A (2013) Angiogenesis and anti-angiogenic therapy in prostate cancer. Crit Rev Oncol Hematol 87:122–131

    PubMed  Google Scholar 

  • Nogueira Pangrazi E, da Silva RF, Kido LA, Montico F, Cagnon VHA (2018) Nintedanib treatment delays prostate dorsolateral lobe cancer progression in the TRAMP model: contribution to the epithelial-stromal interaction balance. Cell Biol Int 42:153–168

    CAS  PubMed  Google Scholar 

  • Noh EM, Park YJ, Kim JM, Kim MS, Kim HR, Song HK, Hong OY, So HS, Yang SH, Kim JS, Park SH, Youn HJ, You YO, Choi KB, Kwon KB, Lee YR (2015) Fisetin regulates TPA-induced breast cell invasion by suppressing matrix metalloproteinase-9 activation via the PKC/ROS/MAPK pathways. Eur J Pharmacol 764:79–86

    CAS  PubMed  Google Scholar 

  • Pal SK, Vuong W, Zhang W, Deng J, Liu X, Carmichael C, Ruel N, Pinnamaneni M, Twardowski P, Lau C, Yu H, Figlin RA, Agarwal N, Jones JO (2015) Clinical and translational assessment of VEGFR1 as a mediator of the premetastatic niche in high-risk localized prostate cancer. Mol Cancer Ther 14:2896–2900

    CAS  PubMed  Google Scholar 

  • Polnaszek N, Kwabi-Addo B, Peterson LE, Ozen M, Greenberg NM, Ortega S, Basilico C, Ittmann M (2003) Fibroblast growth factor 2 promotes tumor progression in an autochthonous mouse model of prostate cancer. Cancer Res 63:5754–5760

    CAS  PubMed  Google Scholar 

  • Ruggeri B, Singh J, Gingrich D, Angeles T, Albom M, Yang S, Chang H, Robinson C, Hunter K, Dobrzanski P, Jones-Bolin S, Pritchard S, Aimone L, Klein-Szanto A, Herbert JM, Bono F, Schaeffer P, Casellas P, Bourie B, Pili R, Isaacs J, Ator M, Hudkins R, Vaught J, Mallamo J, Dionne C (2003) CEP-7055: a novel, orally active pan inhibitor of vascular endothelial growth factor receptor tyrosine kinases with potent antiangiogenic activity and antitumor efficacy in preclinical models. Cancer Res 63:5978–5991

    CAS  PubMed  Google Scholar 

  • Sahadevan K, Darby S, Leung HY, Mathers ME, Robson CN, Gnanapragasam VJ (2007) Selective over-expression of fibroblast growth factor receptors 1 and 4 in clinical prostate cancer. J Pathol 213:82–90

    CAS  PubMed  Google Scholar 

  • Siegel RL, Miller KD, Jemal A (2018) Cancer statistics, 2018. CA Cancer J Clin 68:7–30

    PubMed  Google Scholar 

  • Steinemann G, Jacobsen C, Gerwing M, Hauschild J, von Amsberg G, Hopfner M, Nitzsche B, Honecker F (2016) Activity of nintedanib in germ cell tumors. Anti-Cancer Drugs 27:89–98

    CAS  PubMed  Google Scholar 

  • Weibel ER (1963) Principles and methods for the morphometric study of the lung and other organs. Lab Invest 12:131–155

    CAS  PubMed  Google Scholar 

  • Xu D, Wang X, Lou Y (2017) Association of endothelin-1 gene single-nucleotide polymorphisms and haplotypes with risk of hormone refractory prostate cancer. Pharmazie 72:103–106

    CAS  PubMed  Google Scholar 

  • Yamaguchi K, Izaki H, Takahashi M, Fukumori T, Nishitani M, Sutou Y, Uema K, Kawano A, Hamao T, Kanayama HO (2014) Changes in levels of prostate-specific antigen and testosterone following discontinuation of long-term hormone therapy for non-metastatic prostate cancer. J Med Investig 61:35–40

    Google Scholar 

  • Zar J (1999) Biostatistical analysis. Prentice Hall, New Jersey

    Google Scholar 

Download references

Acknowledgments

Not applicable.

Funding

This work was supported by a grant from the São Paulo Research Foundation (FAPESP 2013/26677-7) and in part by the NCI R01 grant CA195708 (to RA).

Author information

Authors and Affiliations

Authors

Contributions

Study design, collection, analyses and interpretation of data and writing of manuscript: Raquel Frenedoso da Silva and Valéria Helena Alves Cagnon. Collection and analyses of data: Thais Petrochelli Banzato and Letícia Ferreira Alves. Analyses and interpretation of data: Rajesh Agarwal and João Ernesto Carvalho.

Corresponding author

Correspondence to Valéria Helena Alves Cagnon.

Ethics declarations

Conflict of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of the research reported.

Ethical approval

The Ethics Committee on Animal Use (CEUA-UNICAMP) approved this study under protocol number 3285-1, carried out in agreement with the Ethical Principles for Animal Research established by the Brazilian College for Animal Experimentation (COBEA).

Additional information

Publisher’s note

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

Electronic supplementary material

Fig. S1

Experimental design: Animals received treatment from 8 to 12 weeks of age and were euthanized at the end of the treatment (TC12 and TN12, respectively) or at 22 weeks of age (TC22(8–12) and TN22(8–12), respectively). Furthermore, groups receiving treatment from 12 to 16 weeks of age were euthanized either at the end of the treatment (TC16 and TN16, respectively) or at 22 weeks of age (TC22(12–16) and TN22(12–16), respectively). Nintedanib was administered in a dose of 10 mg/kg/day, orally. (JPG 2721 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

da Silva, R.F., Banzato, T.P., Alves, L.F. et al. Antiangiogenic therapy with Nintedanib affects hypoxia, angiogenesis and apoptosis in the ventral prostate of TRAMP animals. Cell Tissue Res 379, 407–420 (2020). https://doi.org/10.1007/s00441-019-03091-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00441-019-03091-x

Keywords

Navigation