Skip to main content
Top
Published in: Journal of Experimental & Clinical Cancer Research 1/2015

Open Access 01-12-2015 | Research article

Halofuginone inhibits phosphorylation of SMAD-2 reducing angiogenesis and leukemia burden in an acute promyelocytic leukemia mouse model

Authors: Patricia A. Assis, Lorena L. De Figueiredo-Pontes, Ana Silvia G. Lima, Vitor Leão, Larissa A. Cândido, Carolina T. Pintão, Aglair B. Garcia, Fabiano P. Saggioro, Rodrigo A Panepucci, Fernando Chahud, Arnon Nagler, Roberto P. Falcão, Eduardo M. Rego

Published in: Journal of Experimental & Clinical Cancer Research | Issue 1/2015

Login to get access

Abstract

Background

Halofuginone (HF) is a low-molecular-weight alkaloid that has been demonstrated to interfere with Metalloproteinase-2 (MMP-2) and Tumor Growth Factor-β (TGF-β) function and, to present antiangiogenic, antiproliferative and proapoptotic properties in several solid tumor models. Based on the fact that high levels of Vascular Endothelial Growth Factor (VEGF) and increased angiogenesis have been described in acute myeloid leukemia and associated with disease progression, we studied the in vivo effects of HF using an Acute Promyelocytic Leukemia (APL) mouse model.

Methods

NOD/SCID mice were transplanted with leukemic cells from hCG-PML/RARA transgenic mice (TM) and treated with HF 150 μg/kg/day for 21 days. The leukemic infiltration and the percentage of VEGF+ cells were evaluated by morphology and flow cytometry. The effect of HF on the gene expression of several pro- and antiangiogenic factors, phosphorylation of SMAD2 and VEGF secretion was assessed in vitro using NB4 and HUVEC cells.

Results

HF treatment resulted in hematological remission with decreased accumulation of immature cell and lower amounts of VEGF in BM of leukemic mice. In vitro, HF modulated gene expression of several pro- and antiangiogenic factors, reduced VEGF secretion and phosphorylation of SMAD2, blocking TGF-β-signaling.

Conclusion

Taken together, our results demonstrate that HF inhibits SMAD2 signaling and reduces leukemia growth and angiogenesis.
Appendix
Available only for authorised users
Literature
1.
go back to reference De Figueiredo-Pontes LL, Assis PA, Santana-Lemos BA, Jacomo RH, Lima AS, Garcia AB, et al. Halofuginone has anti-proliferative effects in acute promyelocytic leukemia by modulating the transforming growth factor beta signaling pathway. PLoS One. 2011;6:e26713.PubMedCentralPubMedCrossRef De Figueiredo-Pontes LL, Assis PA, Santana-Lemos BA, Jacomo RH, Lima AS, Garcia AB, et al. Halofuginone has anti-proliferative effects in acute promyelocytic leukemia by modulating the transforming growth factor beta signaling pathway. PLoS One. 2011;6:e26713.PubMedCentralPubMedCrossRef
2.
go back to reference Chu TL, Guan Q, Nguan CY, Du C. Halofuginone suppresses T cell proliferation by blocking proline uptake and inducing cell apoptosis. Int Immunopharmacol. 2013;16:414–23.PubMedCrossRef Chu TL, Guan Q, Nguan CY, Du C. Halofuginone suppresses T cell proliferation by blocking proline uptake and inducing cell apoptosis. Int Immunopharmacol. 2013;16:414–23.PubMedCrossRef
3.
go back to reference Leiba M, Jakubikova J, Klippel S, Mitsiades CS, Hideshima T, Tai YT, et al. Halofuginone inhibits multiple myeloma growth in vitro and in vivo and enhances cytotoxicity of conventional and novel agents. Br J Haematol. 2012;157:718–31.PubMedCentralPubMedCrossRef Leiba M, Jakubikova J, Klippel S, Mitsiades CS, Hideshima T, Tai YT, et al. Halofuginone inhibits multiple myeloma growth in vitro and in vivo and enhances cytotoxicity of conventional and novel agents. Br J Haematol. 2012;157:718–31.PubMedCentralPubMedCrossRef
4.
go back to reference Abramovitch R, Dafni H, Neeman M, Nagler A, Pines M. Inhibition of neovascularization and tumor growth, and facilitation of wound repair, by halofuginone, an inhibitor of collagen type I synthesis. Neoplasia. 1999;1:321–9.PubMedCentralPubMedCrossRef Abramovitch R, Dafni H, Neeman M, Nagler A, Pines M. Inhibition of neovascularization and tumor growth, and facilitation of wound repair, by halofuginone, an inhibitor of collagen type I synthesis. Neoplasia. 1999;1:321–9.PubMedCentralPubMedCrossRef
5.
go back to reference Elkin M, Miao HQ, Nagler A, Aingorn E, Reich R, Hemo I, et al. Halofuginone: a potent inhibitor of critical steps in angiogenesis progression. FASEB J. 2000;14:2477–85.PubMedCrossRef Elkin M, Miao HQ, Nagler A, Aingorn E, Reich R, Hemo I, et al. Halofuginone: a potent inhibitor of critical steps in angiogenesis progression. FASEB J. 2000;14:2477–85.PubMedCrossRef
6.
go back to reference Abramovitch R, Itzik A, Harel H, Nagler A, Vlodavsky I, Siegal T. Halofuginone inhibits angiogenesis and growth in implanted metastatic rat brain tumor model–an MRI study. Neoplasia. 2004;6:480–9.PubMedCentralPubMedCrossRef Abramovitch R, Itzik A, Harel H, Nagler A, Vlodavsky I, Siegal T. Halofuginone inhibits angiogenesis and growth in implanted metastatic rat brain tumor model–an MRI study. Neoplasia. 2004;6:480–9.PubMedCentralPubMedCrossRef
8.
go back to reference Jakovljevic G, Culic S, Stepan J, Bonevski A, Seiwerth S. Vascular endothelial growth factor in children with neuroblastoma: a retrospective analysis. J Exp Clin Cancer Res. 2009;28:143.PubMedCentralPubMedCrossRef Jakovljevic G, Culic S, Stepan J, Bonevski A, Seiwerth S. Vascular endothelial growth factor in children with neuroblastoma: a retrospective analysis. J Exp Clin Cancer Res. 2009;28:143.PubMedCentralPubMedCrossRef
9.
10.
go back to reference Rajkumar SV, Witzig TE. A review of angiogenesis and antiangiogenic therapy with thalidomide in multiple myeloma. Cancer Treat Rev. 2000;26:351–62.PubMedCrossRef Rajkumar SV, Witzig TE. A review of angiogenesis and antiangiogenic therapy with thalidomide in multiple myeloma. Cancer Treat Rev. 2000;26:351–62.PubMedCrossRef
11.
go back to reference Jorgensen JM, Sorensen FB, Bendix K, Nielsen JL, Olsen ML, Funder AM, et al. Angiogenesis in non-Hodgkin’s lymphoma: clinico-pathological correlations and prognostic significance in specific subtypes. Leuk Lymphoma. 2007;48:584–95.PubMedCrossRef Jorgensen JM, Sorensen FB, Bendix K, Nielsen JL, Olsen ML, Funder AM, et al. Angiogenesis in non-Hodgkin’s lymphoma: clinico-pathological correlations and prognostic significance in specific subtypes. Leuk Lymphoma. 2007;48:584–95.PubMedCrossRef
12.
go back to reference Padro T, Ruiz S, Bieker R, Burger H, Steins M, Kienast J, et al. Increased angiogenesis in the bone marrow of patients with acute myeloid leukemia. Blood. 2000;95:2637–44.PubMed Padro T, Ruiz S, Bieker R, Burger H, Steins M, Kienast J, et al. Increased angiogenesis in the bone marrow of patients with acute myeloid leukemia. Blood. 2000;95:2637–44.PubMed
13.
go back to reference Hatfield KJ, Olsnes AM, Gjertsen BT, Bruserud O. Antiangiogenic therapy in acute myelogenous leukemia: targeting of vascular endothelial growth factor and interleukin 8 as possible antileukemic strategies. Curr Cancer Drug Targets. 2005;5:229–48.PubMedCrossRef Hatfield KJ, Olsnes AM, Gjertsen BT, Bruserud O. Antiangiogenic therapy in acute myelogenous leukemia: targeting of vascular endothelial growth factor and interleukin 8 as possible antileukemic strategies. Curr Cancer Drug Targets. 2005;5:229–48.PubMedCrossRef
14.
go back to reference Aguayo A, Kantarjian HM, Estey EH, Giles FJ, Verstovsek S, Manshouri T, et al. Plasma vascular endothelial growth factor levels have prognostic significance in patients with acute myeloid leukemia but not in patients with myelodysplastic syndromes. Cancer. 2002;95:1923–30.PubMedCrossRef Aguayo A, Kantarjian HM, Estey EH, Giles FJ, Verstovsek S, Manshouri T, et al. Plasma vascular endothelial growth factor levels have prognostic significance in patients with acute myeloid leukemia but not in patients with myelodysplastic syndromes. Cancer. 2002;95:1923–30.PubMedCrossRef
15.
go back to reference Kini AR, Peterson LA, Tallman MS, Lingen MW. Angiogenesis in acute promyelocytic leukemia: induction by vascular endothelial growth factor and inhibition by all-trans retinoic acid. Blood. 2001;97:3919–24.PubMedCrossRef Kini AR, Peterson LA, Tallman MS, Lingen MW. Angiogenesis in acute promyelocytic leukemia: induction by vascular endothelial growth factor and inhibition by all-trans retinoic acid. Blood. 2001;97:3919–24.PubMedCrossRef
16.
go back to reference Alimoghaddam K, Shariftabrizi A, Tavangar SM, Sanaat Z, Rostami S, Jahani M, et al. Anti-leukemic and anti-angiogenesis efficacy of arsenic trioxide in new cases of acute promyelocytic leukemia. Leuk Lymphoma. 2006;47:81–8.PubMedCrossRef Alimoghaddam K, Shariftabrizi A, Tavangar SM, Sanaat Z, Rostami S, Jahani M, et al. Anti-leukemic and anti-angiogenesis efficacy of arsenic trioxide in new cases of acute promyelocytic leukemia. Leuk Lymphoma. 2006;47:81–8.PubMedCrossRef
17.
go back to reference Estey E, Levine RL, Lowenberg B. Current challenges in clinical development of “targeted therapies”: the case of acute myeloid leukemia. Blood. 2015;125:2461–6.PubMedCrossRef Estey E, Levine RL, Lowenberg B. Current challenges in clinical development of “targeted therapies”: the case of acute myeloid leukemia. Blood. 2015;125:2461–6.PubMedCrossRef
18.
go back to reference Estey EH, Giles FJ, Kantarjian H, O’Brien S, Cortes J, Freireich EJ, et al. Molecular remissions induced by liposomal-encapsulated all-trans retinoic acid in newly diagnosed acute promyelocytic leukemia. Blood. 1999;94:2230–5.PubMed Estey EH, Giles FJ, Kantarjian H, O’Brien S, Cortes J, Freireich EJ, et al. Molecular remissions induced by liposomal-encapsulated all-trans retinoic acid in newly diagnosed acute promyelocytic leukemia. Blood. 1999;94:2230–5.PubMed
19.
go back to reference Lo-Coco F, Avvisati G, Vignetti M, Thiede C, Orlando SM, Iacobelli S, et al. Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N Engl J Med. 2013;369:111–21.PubMedCrossRef Lo-Coco F, Avvisati G, Vignetti M, Thiede C, Orlando SM, Iacobelli S, et al. Retinoic acid and arsenic trioxide for acute promyelocytic leukemia. N Engl J Med. 2013;369:111–21.PubMedCrossRef
20.
go back to reference Mathews V, Desire S, George B, Lakshmi KM, Rao JG, Viswabandya A, et al. Hepatotoxicity profile of single agent arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia, its impact on clinical outcome and the effect of genetic polymorphisms on the incidence of hepatotoxicity. Leukemia. 2006;20:881–3.PubMedCrossRef Mathews V, Desire S, George B, Lakshmi KM, Rao JG, Viswabandya A, et al. Hepatotoxicity profile of single agent arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia, its impact on clinical outcome and the effect of genetic polymorphisms on the incidence of hepatotoxicity. Leukemia. 2006;20:881–3.PubMedCrossRef
21.
go back to reference Ribeiro RC, Rego E. Management of APL in developing countries: epidemiology, challenges and opportunities for international collaboration. In: Hematology / the Education Program of the American Society of Hematology American Society of Hematology Education Program. 2006. p. 162–8. Ribeiro RC, Rego E. Management of APL in developing countries: epidemiology, challenges and opportunities for international collaboration. In: Hematology / the Education Program of the American Society of Hematology American Society of Hematology Education Program. 2006. p. 162–8.
22.
go back to reference Mathews V, George B, Chendamarai E, Lakshmi KM, Desire S, Balasubramanian P, et al. Single-agent arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia: long-term follow-up data. J Clin Oncol. 2010;28:3866–71.PubMedCrossRef Mathews V, George B, Chendamarai E, Lakshmi KM, Desire S, Balasubramanian P, et al. Single-agent arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia: long-term follow-up data. J Clin Oncol. 2010;28:3866–71.PubMedCrossRef
23.
go back to reference Tallman MS. Treatment of relapsed or refractory acute promyelocytic leukemia. Best Pract Res Clin Haematol. 2007;20:57–65.PubMedCrossRef Tallman MS. Treatment of relapsed or refractory acute promyelocytic leukemia. Best Pract Res Clin Haematol. 2007;20:57–65.PubMedCrossRef
24.
go back to reference Elkin M, Reich R, Nagler A, Aingorn E, Pines M, De-Groot N, et al. Inhibition of matrix metalloproteinase-2 expression and bladder carcinoma metastasis by halofuginone. Clin Cancer Res. 1999;5:1982–8.PubMed Elkin M, Reich R, Nagler A, Aingorn E, Pines M, De-Groot N, et al. Inhibition of matrix metalloproteinase-2 expression and bladder carcinoma metastasis by halofuginone. Clin Cancer Res. 1999;5:1982–8.PubMed
25.
go back to reference Jordan MC, Zeplin PH. Local inhibition of angiogenesis by halofuginone coated silicone materials. J Mater Sci Mater Med. 2012;23:1203–10.PubMedCrossRef Jordan MC, Zeplin PH. Local inhibition of angiogenesis by halofuginone coated silicone materials. J Mater Sci Mater Med. 2012;23:1203–10.PubMedCrossRef
26.
go back to reference Lin HK, Bergmann S, Pandolfi PP. Cytoplasmic PML function in TGF-beta signalling. Nature. 2004;431:205–11.PubMedCrossRef Lin HK, Bergmann S, Pandolfi PP. Cytoplasmic PML function in TGF-beta signalling. Nature. 2004;431:205–11.PubMedCrossRef
27.
go back to reference Pepper MS. Transforming growth factor-beta: vasculogenesis, angiogenesis, and vessel wall integrity. Cytokine Growth Factor Rev. 1997;8:21–43.PubMedCrossRef Pepper MS. Transforming growth factor-beta: vasculogenesis, angiogenesis, and vessel wall integrity. Cytokine Growth Factor Rev. 1997;8:21–43.PubMedCrossRef
28.
go back to reference Semenza GL. Expression of hypoxia-inducible factor 1: mechanisms and consequences. Biochem Pharmacol. 2000;59:47–53.PubMedCrossRef Semenza GL. Expression of hypoxia-inducible factor 1: mechanisms and consequences. Biochem Pharmacol. 2000;59:47–53.PubMedCrossRef
29.
go back to reference Xavier S, Piek E, Fujii M, Javelaud D, Mauviel A, Flanders KC, et al. Amelioration of radiation-induced fibrosis: inhibition of transforming growth factor-beta signaling by halofuginone. J Biol Chem. 2004;279:15167–76.PubMedCrossRef Xavier S, Piek E, Fujii M, Javelaud D, Mauviel A, Flanders KC, et al. Amelioration of radiation-induced fibrosis: inhibition of transforming growth factor-beta signaling by halofuginone. J Biol Chem. 2004;279:15167–76.PubMedCrossRef
30.
go back to reference Sheffer Y, Leon O, Pinthus JH, Nagler A, Mor Y, Genin O, et al. Inhibition of fibroblast to myofibroblast transition by halofuginone contributes to the chemotherapy-mediated antitumoral effect. Mol Cancer Ther. 2007;6:570–7.PubMedCrossRef Sheffer Y, Leon O, Pinthus JH, Nagler A, Mor Y, Genin O, et al. Inhibition of fibroblast to myofibroblast transition by halofuginone contributes to the chemotherapy-mediated antitumoral effect. Mol Cancer Ther. 2007;6:570–7.PubMedCrossRef
31.
go back to reference Taras D, Blanc JF, Rullier A, Dugot-Senant N, Laurendeau I, Bieche I, et al. Halofuginone suppresses the lung metastasis of chemically induced hepatocellular carcinoma in rats through MMP inhibition. Neoplasia. 2006;8:312–8.PubMedCentralPubMedCrossRef Taras D, Blanc JF, Rullier A, Dugot-Senant N, Laurendeau I, Bieche I, et al. Halofuginone suppresses the lung metastasis of chemically induced hepatocellular carcinoma in rats through MMP inhibition. Neoplasia. 2006;8:312–8.PubMedCentralPubMedCrossRef
32.
go back to reference Nagler A, Ohana M, Shibolet O, Shapira MY, Alper R, Vlodavsky I, et al. Suppression of hepatocellular carcinoma growth in mice by the alkaloid coccidiostat halofuginone. Eur J Cancer. 2004;40:1397–403.PubMedCrossRef Nagler A, Ohana M, Shibolet O, Shapira MY, Alper R, Vlodavsky I, et al. Suppression of hepatocellular carcinoma growth in mice by the alkaloid coccidiostat halofuginone. Eur J Cancer. 2004;40:1397–403.PubMedCrossRef
33.
go back to reference He LZ, Tribioli C, Rivi R, Peruzzi D, Pelicci PG, Soares V, et al. Acute leukemia with promyelocytic features in PML/RARalpha transgenic mice. Proc Natl Acad Sci U S A. 1997;94:5302–7.PubMedCentralPubMedCrossRef He LZ, Tribioli C, Rivi R, Peruzzi D, Pelicci PG, Soares V, et al. Acute leukemia with promyelocytic features in PML/RARalpha transgenic mice. Proc Natl Acad Sci U S A. 1997;94:5302–7.PubMedCentralPubMedCrossRef
34.
go back to reference Lanotte M, Martin-Thouvenin V, Najman S, Balerini P, Valensi F, Berger R. NB4, a maturation inducible cell line with t(15;17) marker isolated from a human acute promyelocytic leukemia (M3). Blood. 1991;77:1080–6.PubMed Lanotte M, Martin-Thouvenin V, Najman S, Balerini P, Valensi F, Berger R. NB4, a maturation inducible cell line with t(15;17) marker isolated from a human acute promyelocytic leukemia (M3). Blood. 1991;77:1080–6.PubMed
35.
go back to reference Wang YH, Dong YY, Wang WM, Xie XY, Wang ZM, Chen RX, et al. Vascular endothelial cells facilitated HCC invasion and metastasis through the Akt and NF-kappaB pathways induced by paracrine cytokines. J Exp Clin Cancer Res. 2013;32:51.PubMedCentralPubMedCrossRef Wang YH, Dong YY, Wang WM, Xie XY, Wang ZM, Chen RX, et al. Vascular endothelial cells facilitated HCC invasion and metastasis through the Akt and NF-kappaB pathways induced by paracrine cytokines. J Exp Clin Cancer Res. 2013;32:51.PubMedCentralPubMedCrossRef
36.
go back to reference Kunkel SL, Campbell Jr DA, Chensue SW, Higashi GI. Species-dependent regulation of monocyte/macrophage Ia antigen expression and antigen presentation by prostaglandin E. Cell Immunol. 1986;97:140–5.PubMedCrossRef Kunkel SL, Campbell Jr DA, Chensue SW, Higashi GI. Species-dependent regulation of monocyte/macrophage Ia antigen expression and antigen presentation by prostaglandin E. Cell Immunol. 1986;97:140–5.PubMedCrossRef
37.
go back to reference Kogan SC, Ward JM, Anver MR, Berman JJ, Brayton C, Cardiff RD, et al. Bethesda proposals for classification of nonlymphoid hematopoietic neoplasms in mice. Blood. 2002;100:238–45.PubMedCrossRef Kogan SC, Ward JM, Anver MR, Berman JJ, Brayton C, Cardiff RD, et al. Bethesda proposals for classification of nonlymphoid hematopoietic neoplasms in mice. Blood. 2002;100:238–45.PubMedCrossRef
38.
go back to reference Rego EM, Ruggero D, Tribioli C, Cattoretti G, Kogan S, Redner RL, et al. Leukemia with distinct phenotypes in transgenic mice expressing PML/RAR alpha, PLZF/RAR alpha or NPM/RAR alpha. Oncogene. 2006;25:1974–9.PubMedCrossRef Rego EM, Ruggero D, Tribioli C, Cattoretti G, Kogan S, Redner RL, et al. Leukemia with distinct phenotypes in transgenic mice expressing PML/RAR alpha, PLZF/RAR alpha or NPM/RAR alpha. Oncogene. 2006;25:1974–9.PubMedCrossRef
39.
40.
go back to reference Santana BA, Pintao MC, Lima RS AE, Scheucher PS, Santos GA, Garcia AB, et al. Asynchronous expression of myeloid antigens in leukemic cells in a PML/RARalpha transgenic mouse model. Braz J Med Biol Res. 2006;39:615–20.PubMedCrossRef Santana BA, Pintao MC, Lima RS AE, Scheucher PS, Santos GA, Garcia AB, et al. Asynchronous expression of myeloid antigens in leukemic cells in a PML/RARalpha transgenic mouse model. Braz J Med Biol Res. 2006;39:615–20.PubMedCrossRef
41.
go back to reference Dong X, Han ZC, Yang R. Angiogenesis and antiangiogenic therapy in hematologic malignancies. Crit Rev Oncol Hematol. 2007;62:105–18.PubMedCrossRef Dong X, Han ZC, Yang R. Angiogenesis and antiangiogenic therapy in hematologic malignancies. Crit Rev Oncol Hematol. 2007;62:105–18.PubMedCrossRef
42.
go back to reference Elkin M, Ariel I, Miao HQ, Nagler A, Pines M, De-Groot N, et al. Inhibition of bladder carcinoma angiogenesis, stromal support, and tumor growth by halofuginone. Cancer Res. 1999;59:4111–8.PubMed Elkin M, Ariel I, Miao HQ, Nagler A, Pines M, De-Groot N, et al. Inhibition of bladder carcinoma angiogenesis, stromal support, and tumor growth by halofuginone. Cancer Res. 1999;59:4111–8.PubMed
43.
go back to reference Ohayon O, Mawasi N, Pevzner A, Tryvitz A, Gildor T, Pines M, et al. Halofuginone upregulates the expression of heparanase in thioacetamide-induced liver fibrosis in rats. Lab Investig: J Tech Meth Pathol. 2008;88:627–33.CrossRef Ohayon O, Mawasi N, Pevzner A, Tryvitz A, Gildor T, Pines M, et al. Halofuginone upregulates the expression of heparanase in thioacetamide-induced liver fibrosis in rats. Lab Investig: J Tech Meth Pathol. 2008;88:627–33.CrossRef
44.
45.
go back to reference Schuch G, Machluf M, Bartsch Jr G, Nomi M, Richard H, Atala A, et al. In vivo administration of vascular endothelial growth factor (VEGF) and its antagonist, soluble neuropilin-1, predicts a role of VEGF in the progression of acute myeloid leukemia in vivo. Blood. 2002;100:4622–8.PubMedCrossRef Schuch G, Machluf M, Bartsch Jr G, Nomi M, Richard H, Atala A, et al. In vivo administration of vascular endothelial growth factor (VEGF) and its antagonist, soluble neuropilin-1, predicts a role of VEGF in the progression of acute myeloid leukemia in vivo. Blood. 2002;100:4622–8.PubMedCrossRef
46.
go back to reference Oka Y, Tashiro H, Shirasaki R, Sugao T, Nishi R, Akiyama N, et al. Vascular endothelial growth factor acted as autocrine growth factor in an acute promyelocytic leukemia case. Leuk Lymphoma. 2010;51:717–9.PubMedCrossRef Oka Y, Tashiro H, Shirasaki R, Sugao T, Nishi R, Akiyama N, et al. Vascular endothelial growth factor acted as autocrine growth factor in an acute promyelocytic leukemia case. Leuk Lymphoma. 2010;51:717–9.PubMedCrossRef
47.
go back to reference Ossenkoppele GJ, Stussi G, Maertens J, Van Montfort K, Biemond BJ, Breems D, et al. Addition of bevacizumab to chemotherapy in acute myeloid leukemia at older age: a randomized phase 2 trial of the Dutch-Belgian Cooperative Trial Group for Hemato-Oncology (HOVON) and the Swiss Group for Clinical Cancer Research (SAKK). Blood. 2012;120:4706–11.PubMedCrossRef Ossenkoppele GJ, Stussi G, Maertens J, Van Montfort K, Biemond BJ, Breems D, et al. Addition of bevacizumab to chemotherapy in acute myeloid leukemia at older age: a randomized phase 2 trial of the Dutch-Belgian Cooperative Trial Group for Hemato-Oncology (HOVON) and the Swiss Group for Clinical Cancer Research (SAKK). Blood. 2012;120:4706–11.PubMedCrossRef
48.
49.
go back to reference Du R, Lu KV, Petritsch C, Liu P, Ganss R, Passegue E, et al. HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell. 2008;13:206–20.PubMedCentralPubMedCrossRef Du R, Lu KV, Petritsch C, Liu P, Ganss R, Passegue E, et al. HIF1alpha induces the recruitment of bone marrow-derived vascular modulatory cells to regulate tumor angiogenesis and invasion. Cancer Cell. 2008;13:206–20.PubMedCentralPubMedCrossRef
50.
go back to reference Chen P, Yang LL, Yang HS, Wang YS, Li G, Wu Y, et al. Synergistic antitumor effect of CXCL10 with hyperthermia. J Cancer Res Clin Oncol. 2008;134:679–87.PubMedCrossRef Chen P, Yang LL, Yang HS, Wang YS, Li G, Wu Y, et al. Synergistic antitumor effect of CXCL10 with hyperthermia. J Cancer Res Clin Oncol. 2008;134:679–87.PubMedCrossRef
51.
go back to reference Scrideli CA, Cortez MA, Yunes JA, Queiroz RG, Valera ET, Da Mata JF, et al. mRNA expression of matrix metalloproteinases (MMPs) 2 and 9 and tissue inhibitor of matrix metalloproteinases (TIMPs) 1 and 2 in childhood acute lymphoblastic leukemia: potential role of TIMP1 as an adverse prognostic factor. Leuk Res. 2010;34:32–7.PubMedCrossRef Scrideli CA, Cortez MA, Yunes JA, Queiroz RG, Valera ET, Da Mata JF, et al. mRNA expression of matrix metalloproteinases (MMPs) 2 and 9 and tissue inhibitor of matrix metalloproteinases (TIMPs) 1 and 2 in childhood acute lymphoblastic leukemia: potential role of TIMP1 as an adverse prognostic factor. Leuk Res. 2010;34:32–7.PubMedCrossRef
52.
go back to reference Gavish Z, Pinthus JH, Barak V, Ramon J, Nagler A, Eshhar Z, et al. Growth inhibition of prostate cancer xenografts by halofuginone. Prostate. 2002;51:73–83.PubMedCrossRef Gavish Z, Pinthus JH, Barak V, Ramon J, Nagler A, Eshhar Z, et al. Growth inhibition of prostate cancer xenografts by halofuginone. Prostate. 2002;51:73–83.PubMedCrossRef
53.
go back to reference Grudzien MM, Low PS, Manning PC, Arredondo M, Belton Jr RJ, Nowak RA. The antifibrotic drug halofuginone inhibits proliferation and collagen production by human leiomyoma and myometrial smooth muscle cells. Fertil Steril. 2010;93:1290–8.PubMedCentralPubMedCrossRef Grudzien MM, Low PS, Manning PC, Arredondo M, Belton Jr RJ, Nowak RA. The antifibrotic drug halofuginone inhibits proliferation and collagen production by human leiomyoma and myometrial smooth muscle cells. Fertil Steril. 2010;93:1290–8.PubMedCentralPubMedCrossRef
54.
go back to reference McGaha TL, Phelps RG, Spiera H, Bona C. Halofuginone, an inhibitor of type-I collagen synthesis and skin sclerosis, blocks transforming-growth-factor-beta-mediated Smad3 activation in fibroblasts. J Invest Dermatol. 2002;118:461–70.PubMedCrossRef McGaha TL, Phelps RG, Spiera H, Bona C. Halofuginone, an inhibitor of type-I collagen synthesis and skin sclerosis, blocks transforming-growth-factor-beta-mediated Smad3 activation in fibroblasts. J Invest Dermatol. 2002;118:461–70.PubMedCrossRef
55.
go back to reference Roffe S, Hagai Y, Pines M, Halevy O. Halofuginone inhibits Smad3 phosphorylation via the PI3K/Akt and MAPK/ERK pathways in muscle cells: effect on myotube fusion. Exp Cell Res. 2010;316:1061–9.PubMedCrossRef Roffe S, Hagai Y, Pines M, Halevy O. Halofuginone inhibits Smad3 phosphorylation via the PI3K/Akt and MAPK/ERK pathways in muscle cells: effect on myotube fusion. Exp Cell Res. 2010;316:1061–9.PubMedCrossRef
56.
go back to reference Juarez P, Mohammad KS, Yin JJ, Fournier PG, McKenna RC, Davis HW, et al. Halofuginone inhibits the establishment and progression of melanoma bone metastases. Cancer Res. 2012;72:6247–56.PubMedCentralPubMedCrossRef Juarez P, Mohammad KS, Yin JJ, Fournier PG, McKenna RC, Davis HW, et al. Halofuginone inhibits the establishment and progression of melanoma bone metastases. Cancer Res. 2012;72:6247–56.PubMedCentralPubMedCrossRef
57.
go back to reference Kobayashi T, Liu X, Wen FQ, Fang Q, Abe S, Wang XQ, et al. Smad3 mediates TGF-beta1 induction of VEGF production in lung fibroblasts. Biochem Biophys Res Commun. 2005;327:393–8.PubMedCrossRef Kobayashi T, Liu X, Wen FQ, Fang Q, Abe S, Wang XQ, et al. Smad3 mediates TGF-beta1 induction of VEGF production in lung fibroblasts. Biochem Biophys Res Commun. 2005;327:393–8.PubMedCrossRef
58.
go back to reference Jeon SH, Chae BC, Kim HA, Seo GY, Seo DW, Chun GT, et al. Mechanisms underlying TGF-beta1-induced expression of VEGF and Flk-1 in mouse macrophages and their implications for angiogenesis. J Leukoc Biol. 2007;81:557–66.PubMedCrossRef Jeon SH, Chae BC, Kim HA, Seo GY, Seo DW, Chun GT, et al. Mechanisms underlying TGF-beta1-induced expression of VEGF and Flk-1 in mouse macrophages and their implications for angiogenesis. J Leukoc Biol. 2007;81:557–66.PubMedCrossRef
59.
go back to reference Clifford RL, Deacon K, Knox AJ. Novel regulation of vascular endothelial growth factor-A (VEGF-A) by transforming growth factor (beta)1: requirement for Smads, (beta)-CATENIN, AND GSK3(beta). J Biol Chem. 2008;283:35337–53.PubMedCrossRef Clifford RL, Deacon K, Knox AJ. Novel regulation of vascular endothelial growth factor-A (VEGF-A) by transforming growth factor (beta)1: requirement for Smads, (beta)-CATENIN, AND GSK3(beta). J Biol Chem. 2008;283:35337–53.PubMedCrossRef
60.
go back to reference Pepper MS, Vassalli JD, Orci L, Montesano R. Biphasic effect of transforming growth factor-beta 1 on in vitro angiogenesis. Exp Cell Res. 1993;204:356–63.PubMedCrossRef Pepper MS, Vassalli JD, Orci L, Montesano R. Biphasic effect of transforming growth factor-beta 1 on in vitro angiogenesis. Exp Cell Res. 1993;204:356–63.PubMedCrossRef
Metadata
Title
Halofuginone inhibits phosphorylation of SMAD-2 reducing angiogenesis and leukemia burden in an acute promyelocytic leukemia mouse model
Authors
Patricia A. Assis
Lorena L. De Figueiredo-Pontes
Ana Silvia G. Lima
Vitor Leão
Larissa A. Cândido
Carolina T. Pintão
Aglair B. Garcia
Fabiano P. Saggioro
Rodrigo A Panepucci
Fernando Chahud
Arnon Nagler
Roberto P. Falcão
Eduardo M. Rego
Publication date
01-12-2015
Publisher
BioMed Central
Published in
Journal of Experimental & Clinical Cancer Research / Issue 1/2015
Electronic ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-015-0181-2

Other articles of this Issue 1/2015

Journal of Experimental & Clinical Cancer Research 1/2015 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine