Skip to main content
Erschienen 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

verfasst von: 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

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2015

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.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​s13046-015-0181-2) contains supplementary material, which is available to authorized users.
Patricia A. Assis and Lorena L. De Figueiredo-Pontes contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

§PAA and LLFP have equally contributed to the study. PAA, LLFP, NA and EMR: Conceived and designed the experiments; PAA, LLFP, ASGL, VL, LAC, CTP, ASGL and FPS: Performed the experiments; PAA, LLFP, VL, FPS and FC: Analyzed the data; RAP, FC, AN, RPF and EMR: Contributed reagents/materials/analysis tools; PAA and EMR: Wrote the manuscript. All authors read and approved the final manuscript.

Background

Halofuginone (HF) is a low-molecular-weight quinazolinone alkaloid that has been demonstrated to present potent antitumor effects due antiproliferative [1, 2] and proapoptotic [3] properties. Treatment with HF impaired proliferation and induced apoptosis of several multiple myeloma cell lines and showed antitumor activity in a myeloma xenograft mouse model [3]. In addition, HF antiangiogenic properties also have been shown to contribute to control of tumor growth [46]. In a rat brain tumor model, HF treatment markedly reduced vascularization and vessel maturation resulting in reduced tumor burden and metastasis [6].
Angiogenesis is one of the most important mechanisms of tumor progression [7, 8] and has been associated to development of hematological malignancies [9]. Several studies have reported an increase in bone marrow (BM) microvessel density (MVD) and high VEGF (Vascular Endothelial Growth Factor) expression in multiple myeloma [10], non-Hodgkin lymphoma [11], and acute leukemias [12, 13]. Particularly, in acute myeloid leukemia (AML), high levels of VEGF and increased MVD were found in leukemic BM biopsies [12] and correlated with lower rates of complete remission and lower overall survival [14]. In a study analyzing 12 BM biopsies from patients with acute promyelocytic leukemia (APL), Kini et al. [15] detected increased MVD associated with high VEGF expression. After standard treatment with ATRA (all-trans retinoic acid), VEGF levels and BM MVD were normalized. Another clinical study with 17 new cases of APL associated the efficacy of the arsenic trioxide treatment with its antiangiogenic effect, demonstrated by the reduced MVD in the BM and increased survival [16].
Despite the fact that all-trans retinoic acid (ATRA) is largely applied for the treatment of APL, the use of ATRA alone does not achieve long-term remission [17, 18]. Thus, associations of ATRA with chemotherapy or arsenic trioxide have been more successful in improve long-term survival [19]. However, these treatment regiments have limited application for elderly patients or those with serious heart conditions [20]. In addition, the therapy with arsenic trioxide is not available in several developing countries [21] and the 10-20 % of patients treated with ATRA-chemotherapy or ATRA-arsenic trioxide association will relapse [22, 23]. Therefore, the development of new strategies for APL therapeutics is necessary and angiogenesis is a potential target to achieve a better outcome.
The exact mechanism by which HF impairs angiogenesis has not yet been fully characterized. Elkin et al. [24] reported that HF treatment led to the reduction in metalloproteinase-2 (MMP-2) expression and inhibition of bladder carcinoma metastasis in an animal model. However, HF effects may not be specific to malignant tissues, since the implant of silicone coated with HF resulted in reduced expression of MMP-2, bFGF and TGF-β in normal muscle [25]. In APL the TGF-β pathway is deregulated by the cytoplasmic PML isoform (cPML) [26] and we have demonstrated that HF inhibited cell proliferation and induced apoptosis in the APL cell line NB4 through modulation of TGF-β-target genes, such as c-Myc and p21 [1]. In addition, it is noteworthy to point out that the promoter region of the VEGF gene contains DNA-binding sequences for hypoxia-inducible factor 1 alpha (HIF-1α) and for the TGF-β-signaling mediators SMADs [27, 28]. Thus, activation of TGF-β pathway may be associated with increased VEGF expression and angiogenesis in acute leukemias. Here we analyzed the in vivo antileukemic effect of HF and its association with the modulation of proangiogenic and antiangiogenic factors using APL as a bone fide example of acute leukemia with increased angiogenesis.

Methods

Halofuginone

Halofuginone (HF-dl-trans-7-bromo-6-chloro-3-(3-(3-hydroxy-piperidyl)acetonyl)-(3H)-quinazolinone hydrobromide) was solubilized in lactic acid priors to dilute into phosphate buffer solution. In vitro ED50 was previous determined [1] in vivo doses were delineated based on previous reports of the use of this drug in solid tumors and fibrosis models [2932].

Mice and leukemia transplant model

BM angiogenesis was first analyzed in leukemic hCG-PML/RARA transgenic mice (TM). Notably, in this transgenic model, a lethal form of leukemia that closely resembles human APL occurs after a long pre-leukemic phase (12–15 months) and affects only 10–15 % of the TM [33]. To analyze in vivo effects of HF we developed an APL transplanted model. Thus, NOD/SCID mice aged 10 to 12 weeks were irradiated with 250 cGy and 24 hours later, 2 × 106 viable cells from leukemic hCG-PML/RARA transgenic mice (TM) were intravenously injected. Twenty-four hours after transplantation, NOD/SCID mice were then submitted to daily treatment with 150 μg/kg/day HF (n = 5) or vehicle only (0.9 % NaCl; n = 5) as an intraperitoneal injection for 21 consecutive days. Previous experiments were performed to confirm leukemic infiltration in transplanted animals and to test the efficacy and toxicity of HF. All animal experiments using mice were conducted according to national guidelines for the care and use of laboratory animals (Brazilian College of Animal Experimentation) and were approved by the institutional Animal Experimentation Ethics Committee (protocol number 062/2008).

Cell culture

NB4 cells, a permanent cell line harboring PML-RARα t (15;17), characterizing an APL cell line [34], was used for in vitro assays. Briefly, cells were cultured in RPMI 1640 with 10 % fetal calf serum (FCS, Gibco, BRL, UK). Human Umbilical Vein Endothelial Cells (HUVEC) were maintained in EBM medium supplemented with 20 % FCS [35]. All cells were maintained at 37 °C under 5 % CO2.

Quantification of VEGF, angiogenin, and TGF-β

NB4 cells were treated with HF for 72 hours at doses ranging from 25 to 100 ng/mL. VEGF, angiogenin and TGF-β concentrations in supernatants were determinate by CBA (Cytometric Bead Array, BD Biosciences, San Jose, CA, USA) and/or ELISA (BD Biosciences, San Jose, CA, USA), following manufacturer’s recommendations.

Phospho-SMAD2 (P-SMAD2) identification

The phosphorylation of SMAD2 was assessed in total protein extracts obtained from NB4 cells treated with 50, 100, and 200 ng/mL doses of HF for 6, 12, and 24 hours. We used the PathScan® Phospho-Smad2 (Ser465/467) Sandwich ELISA Kit (Cell Signaling, Danvers, MA, USA) that provides the P-SMAD2 level as a direct absorbance reason. To assess the phosphorylated protein, TGF-β1 cytokine (Sigma-Aldrich, St. Louis, MO, USA) was added to the culture (1ng/mL) one hour before the ending of HF treatment.

Real time PCR and PCR array

mRNA expression was assessed by real time PCR. Briefly, total RNA was isolated using Trizol reagent (Invitrogen, Carlsbad, CA, USA) and was reverse transcribed using the cDNA High Capacity Archive kit (Applied Biosystems, Foster City, CA, USA). cDNA samples were used to quantify gene expression using specific primers and probes TaqMan® (Applied Biosystems, Foster City, CA, USA). Gapdh was included as endogenous internal control. PCR array was performed using the angiogenesis RT2 Profiler™ PCR Arrays kit (SA Biosciences, Frederick, MD, USA), in which each cDNA sample was processed in a 96-well plate containing 84 angiogenesis-related genes and B2M, HPRT1, RPL13A, ACTB, and GAPDH were included as endogenous internal controls in all plates. mRNA expression of non-treated cells or non-leukemic mice are expressed as Fold Change = 1. Amplification was performed in plate duplicates in a 7500 Real-Time PCR System (Applied Biosystems, Foster City, CA, USA).

Immunohistochemistry staining for VEGF in bone marrow sections

Vertebra samples were collected from mice, fixed and serial 5-μm sections were prepared in paraffin. Immunohistochemistry was performed with primary anti-VEGF antibodies (Santa Cruz Biotechnology, Santa Cruz, CA, USA) and anti-α-actin (for TM mice) and anti-CD34 (for transplante mice) (Becton Dickinson, San Jose, CA, USA) at 1:500, 1:800 and 1:50, respectively. Peroxidase activity was revealed by 3,3′-diaminobenzidine as chromogen. Alpha-actin and CD34 expression was used for assessing microvessel density (MVD), quantified as the number of microvessels/mm2 after analyzing three high-power fields (400 ×).

Analysis of the hematological responses to HF in vivo

At day 21 mice were sacrificed after being subjected to a cardiac puncture to obtain peripheral blood [36] samples. Age-matched NOD/SCID healthy animals were used as controls (WT; n = 3). Automated counts for white blood cells (WBC) were performed using a T-890 Coulter cell counter (Coulter Corporation, Hialeah, FL, USA), and differential counts were obtained from Leishman-Wright-Giemsa-stained smears. For morphological analysis, 100 cells of PB were counted, and then myeloid cells were classified as immature, intermediate, or mature, according to the Bethesda criteria [37]. Bone marrow (BM) cells were obtained by flushing of the bone cavities and the cells were used for cytospin slide preparation and staining with Leishman-Wright-Giemsa. Imprint slides of the spleen were also performed to quantify the hematological response to HF.

Immnunophenotyping of bone marrow cells

BM cells were characterized by immnunophenotyping using the following combinations of mAbs: CD117-PE/CD16/32-FITC/CD45-Per-CP; CD11b-PE/Gr-1-FITC/CD45-PerCP. Fluorochrome-conjugated isotypic antibodies were used as negative controls. All antibodies were purchased from BD Biosciences Pharmingen (San Diego, CA, USA). A minimum of 10,000 events/tube were acquired with a FACScalibur flow cytometer. Viable CD45-positive cells were gated and the percentage of each cell subset was determined using the CellQuest software (Becton Dickinson).

ChIP assay

NB4 cells were stimulated with Halofuginone (200 ng/mL), TGF-β (50 ng/ml) or Halofuginone (200ng/mL) + TGF-β (50ng/mL), for 6 hours and the chormatin immunoprecipitation was performed with Magna ChIP™ G (Millipore, Billerica, MA, USA) according to the instructions of manufacturer. Briefely, the crosslink was made with 1 % formaldehyde followed by addition of 125mM glicine to quench unreacted formaldehyde. Samples were then resuspended in cell lysis buffer containing Proteases Inhibitors before addition of Nuclear Lysis Buffer. The final lysates were sonicated to shear the genomic DNA into around 200-1000pb fragments. ChIP assay was performed on the supernatants by addition of 5 μg of each antibody: Anti-P-Smad2 (phospho S467 - ab192191); Anti-Smad2 + Smad3 (ab63672); Anti-HIF-1α (ab2185) and IgG rabbit polyclonal (ab27478). Primers for Vegf promoter region were custom designed (Additional file 1: Table S1). Graphics show Fold enrichment over IgG antibodie control for each sample and primer. Primers were purchased from Sigma- Aldrich (St. Louis, MO, USA).

Statistical analysis

All experiments were evaluated by analysis of the variance (ANOVA) between groups followed by the Bonferroni’s correction post-test or Mann-Whitney unpaired t-test. In all comparisons, a significance level of P < 0.05 was considered to be significant. Statistical analyses were performed using SPSS 13.0 software.

Results

Angiogenesis is aberrantly increased in the APL transgenic mouse model

Based on the demonstration of high levels of VEGF in BM biopsies from APL patients [15], we first investigated VEGF expression in an APL transgenic mouse (TM) model expressing the PML/RARα fusion gene under the control of human cathepsin gene promoter (hCG) [33, 38]. Fig. 1a and b show the immunohistochemistry analysis of VEGF expression in leukemic and control BM TM samples. There was a striking increase in the number VEGF positive cells in leukemic BM and, by correlating with H&E staining (not shown), it was possible to identify that most VEGF positive cells were leukemic blasts. In addition, the number of microvessels/mm2 in BM sections of leukemic TM was higher than that of controls (mean of 65.2 ± 38.44 vs. 18.25 ± 7.66 microvessels/mm2, respectively). These results demonstrate that the PML-RARα TM model reproduces the increased MVD and VEGF reported in human APL.

Halofuginone modulates expression of genes associated with angiogenesis

Jordan and Zeplin, showed that HF significantly reduces the gene expression of pro-angiogenic factors in physiologic conditions in muscle cells [25]. In order to evaluate the effect of HF in leukemic cells, we treated the APL cell line NB4 and performed a screening of gene expression using the PCR array method analyzing 84 predefined genes involved in angiogenesis (Additional file 2: Table S2). Among the 40 genes whose expression was modulated by incubation with HF at the dose of 50ng/ml for 6 hours, 16 encoded for growth factors and receptors, 6 for cytokines, 5 for proteases inhibitors and matrix proteins, 4 for adhesion molecules and 3 for transcription factors. As highlighted in Fig. 2a and b the expression of most proangiogenic factors was decreased and the antiangiogenic factors TIMP2 and CXCL10 was increased at all HF concentrations. The largest differences were noted for HIF-1α (mean of 0.48 ± 0.33 fold change), HGF (0.12 ± 0.056), angiopoetin-1 (ANGPT-1: 0.38 ± 0.19), angiopoetin-2 (ANGPT-2: 0.57 ± 0.16), TIMP2 (2.1 ± 0.74) and CXCL10 (16.5 ± 5.4). HF dose dependent effect was detected for VEGFA, TIMP2 and CXCL10.
We next evaluate if the HF effects over gene expression would extend to endothelial cells. Interestingly, HUVEC cells treated with the aforementioned HF concentrations, showed increased VEGF gene expression (9.3 ± 3.34; at 100 ng/mL) and protein secretion (Fig. 3a and 3b). Moreover, ANGPT-1, HIF-1α, HGF, TIMP2 and CXCL10 did not show significant change in gene expression while ANGPT-2 showed increased expression (3.5 ± 1.02; at 200 ng/mL).

Halofuginone inhibits pro-angiogenic mediators in the APL cell line NB4

Previously, we have demonstrated that HF at concentrations higher than 75 ng/ml induced TGF-β and TGFβ-R2 protein expression in NB4 cells [1]. TGF-β-cell responses are coordinated through SMADs phosphorylation, which translocate to nucleus and work as transcription factors [39]. Thus, we further analyzed the phosphorylation of SMAD2 in NB4 cells. HF treatment for 6 and 24 hours impaired phosphorylation of SMAD2 (Fig. 4a), which was a dose- and time-dependent effect that lasted at least for 24 hours. Based on the fact that the promoter region of the VEGF gene contains DNA-binding sequences for SMADs [27, 28] and the previous report that the NB4 cells secretes high levels of VEGF [15], we evaluated the effect of HF treatment in NB4 VEGF production. Quantification of cell-culture supernatants of NB4 cells treated with HF for 72 hours showed that VEGF levels were strongly reduced using HF concentration of 200 ng/mL. (Fig. 4b). In order to verify if SMAD2 has a role as transcription factor for VEGF expression in NB4 cells, we performed a ChIP assay using antibodies for the phosphorylated form of SMAD2 (P-SMAD2) and for the heterodimer SMAD2/3. Analysis of approximately 3,700 pb neighboring the transcription start site (TSS) of VEGF gene showed that, NB4 stimulation with TGF-β increases P-SMAD2 binding in several sites upstream and downstream VEGF. HF treatment prior TGF-β stimulation enhanced P-SMAD2 (Fig. 4c). On the other hand, ChIP assay using anti-SMAD2/3 did not showed binding of unphosphorylated SMADs upon stimulation with TGF-β, however the previous treatment with HF showed increase increase in two sites (2,500 and 3,600 upstream TSS) (Fig. 4d). Similarly, HIF-1α binding was not induced only by TGF-β stimulation, and was enhanced with HF treatment (Additional file 3: Figure S1A). Anti-RNA Polimerase II was used as a control of the ChIP assay (Additional file 3: Figure S1B), and showed increased binding in all the sites analyzed upon TGF-β stimulation with or without HF treatment.

Halofuginone displays an anti-leukemic effect in the in vivo model of APL

To investigate whether the anti-angiogenic properties of HF resulted in anti-leukemic effects in APL, we used a transplant model of PML-RARα leukemic cells in NOD/SCID mice. All recipients developed extensive BM and spleen infiltration, leukocytosis and succumbed of the disease within 30-40 days [1]. This strategy was necessary due to the long latency and low penetrance of the transgenic model, which require the observation of a large cohort of TM. Therefore, recipient mice were treated with HF i.p. for 21 days starting on Day 2 post transplant and at the end of treatment mice were sacrificied. Treatment with HF significantly reduced white blood cell (WBC) counts in leukemic mice (Fig. 5a). [33]. Morphological analysis of Leishman-Wright-Giemsa-stained slides of BM and spleen imprints confirmed the infiltration of immature cells in leukemic animals and showed a significant reduction of this cell pattern in the HF-treated mice (Fig. 5b). The immunophenotypic analysis of BM showed that HF-treated mice had a higher percentage of more differentiated CD11b+/Gr-1+ myeloid cells and lower of CD117+CD16/32+ (previously defined as murine leukemic cells [40]) compared to leukemic non-treated mice (Fig. 5c, d and e). Finally, in addition to the anti-proliferative action, the reduction of leukemia burden observed with HF treatment correlated with antiangiogenic effects. Quantitative and qualitative expression of VEGF by immunohistochemistry showed a reduced number of VEGF+ cells and lower VEGF expression in BM sections from HF-treated mice than in non-treated control (Fig. 6a). Likewise, in our findings with the leukemic PML-RARα TM, most of the VEGF positive cells corresponded to immature cells (Fig. 6b). Nevertheless, the reduction of the MVD in HF-treated mice did not reach statistical significance (158 ± 32.5 vs. 118 ± 35 microvessels/mm2 in controls (n = 5) and treated leukemic mice (n = 5), respectively). In the other hand, PCR analysis of BM samples also showed reduction in VEGF, HIF-1α and ANGPT-2 gene expression in HF-treated mice (Fig. 6c), whereas ANGPT-1, HGF, TIMP2 and CXCL10 had no change in leukemic mice (data not shown).

Discussion

Here we report that HF reduced tumor burden in an APL mouse model as showed by the cytomorphological and immunophenotyping analysis of the BM after 21 days of treatment. This is the first study to show that leukemic hCG-PML/RARA TM had increased VEGF expression in BM, reinforcing that VEGF may be a mediator supporting leukemogenesis [41]. Using a transplant model of PML-RARα leukemic cells, the treatment with HF reduced the number of VEGF+ cells and VEGF gene expression in BM of leukemic mice. However, HF treatment did not result in a significant reduction of MVD. This may be a consequence of the transplant model used, which led to a massive infiltration of the BM with a shorter survival compared to solid tumor models. In this regard it is worthy pointing out that in the study by Elkin et al. the animals with bladder cancer were treated with oral HF for 26 and 29 weeks [42].
In addition, the treatment of the endothelial cell HUVEC with HF did not change the gene expression of ANGPT-1, HIF-1α, TIMP2 and CXCL10, whereas increased VEGF gene expression and protein secretion. These data suggests that HF effects may present variation according to the cell type, which is in accord with the previous demonstrated increase in VEGF production by fibrotic liver cells from rats treated with HF [43]. Furthermore, it is well established that tumor cells-derived VEGF stimulates endothelial cells to produce VEGF [44]. Considering that the basal production of VEGF showed by NB4 cells is over 100-fold higher, the induction of VEGF by HF treatment in endothelial cells may not have impact in the angiogenesis process.
In non-APL AML Schuch et al. [45] demonstrated that VEGF administration resulted in enhanced tumor growth and vascularization whereas treatment with a VEGF antagonist soluble NRP-1 inhibited tumor angiogenesis and growth in a chloroma mouse model. Moreover, these authors showed that the injection of adenovirus encoding NRP-1 reduced the leukemic burden and prolonged the survival of leukemic mice. Recently, Oka et al. described a high level of VEGF-A in the serum of an APL patient at diagnosis and the ex-vivo treatment with an anti-VEGF-A reduced leukemic cell proliferation [46]. Unfortunately, in the phase II HOVON/SAKK AML study, the administration of the anti-VEGF agent bevacizumab along with standard chemotherapy did not result in better outcome in older patients [47]. Thus, despite experimental results indicating that VEGF have a potential therapeutic benefit, the HOVON/SAKK study suggests that inhibition of VEGF may not be sufficient to control angiogenesis or tumor growth.
Considering HF antiangiogenic properties, we used a PCR array to screening angiogenic mediators in the APL cell line NB4. Our data showed a down-regulation mediated by HF in several pro-angiogenic factors, including HFG, HIF-1α, ANGPT-1 and -2, whereas the anti-angiogenic factors TIMP2 and CXCL10 were over expressed. Indeed, HGF is a well-recognized proangiogenic mediator which inhibition reduces tumor growth and metastasis [48], as well HIF-1α, which is associated with poor prognosis in cancer due stimulation of VEGFR+ cells migration to the tumor [49]. Contrary, CXCL10 and TIMP2 had they antiangiogenic potential demonstrated. Mice with fibrosarcoma treated with CXCL10 combined with hypothermia presented reduced tumor burden and MVD [50], whereas the MMP2-inhibitor TIMP2, have been recently described as important prognostic factors in acute lymphoblastic leukemia [51]. In the leukemia mouse model, we did observe increased gene expression of VEGF, HIF-1α and ANGPT-2, which were significantly reduced after HF treatment. These PCR data support that HF antiangiogenic effect implicates multiple targets, as demonstrated by Jordan ans Zeplin [25], MMP-2, bFGF and TGF-β were down-modulated by HF in physiologic conditions.
On the other hand, addition of HF to NB4 cells cultures did not result in morphological differentiation [1] likewise, no differentiation was observed in BM and blood smears of recipient mice at seven and 14 days of HF treatment (data not shown). Therefore, we believe that increase in differentiated cells in BM of HF treated mice reflects the restoration of normal hematopoiesis secondary to the significant reduction of the leukemic infiltration. Together, our results corroborate with previous studies reporting the anti-proliferative and proapoptotic actions of HF in solid tumors [32, 52] and in leiomyoma cell line [53] and with recent observations from our group demonstrating that HF induced the blockade of cell cycle progression at G1/S transition and apoptosis in NB4 cells [1]. Here we showed that NB4 cells treated with HF had SMAD2 phosphorylation abrogated indicating that the TGF-β-signaling was inhibited, which is in agreement with previous studies [5456]. It is proposed that VEGF gene expression is targeted by TGF-β- signaling [5759] and that low concentration of TGF-β cooperate with VEGF to stimulate epithelial cell proliferation and migration [60]. Thus, one possible explanation for the VEGF reduction induced by HF is the inhibition of the TGF-β-signaling. One result suggest that HF acts by decreasing SMAD-2 phosphorylation triggered by TGF-β. However, ChIP analysis showed that, despite reduction of SMAD phosphorylation, HF treatment increases P-SMAD2 binding to VEGF promoter region. Importantly, it has been demonstrated that SMAD2 is not required for VEGF gene expression, and that SMAD3 is the potential transcription factor linking TGF-β-signaling and VEGF [57].
It has been proposed that the PML-RARα fusion protein, similarly to cPML interacts and co-localizes with SMAD2/3 and SARA thus interfering with the phosphorylation and nuclear translocation of this complex Lin et al. [26]. The authors hypothesized that PML/RARα in the cytosol inhibits TGF-β-signaling by sequestering cPML away from the TFβR1/SARA/SMAD2/3 complex thus preventing the trafficking to the early endosome. This inhibition could be further enhanced by interfering with nuclear isoform PML4, which has been demonstrated to transactivate TGF-β target genes. Our results showed that NB4 cells responded to the addition of TGF-β in culture medium with an increase of about 20-fold in SMAD2 phosphorylation. HF was capable of inhibiting TGF-β-induced SMAD2 phosphorylation. Taken together with our previous report that HF blocked NB4 proliferation and reduced the leukemia burden in vivo we postulated that despite the interference of PML/RARα with TFβR1/SARA/SMAD2/3 complex, the TGF-β pathway remain active in APL and contributes for leukemia progression.

Conclusion

Here we showed HF can affect multiply mechanism and that HF treatment reduced proangiogenic factors expression and TGF-β-signaling in vitro, in addition to displaying antiangiogenic and antileukemic effects in an animal model of APL. Considering the importance of angiogenesis and deregulation of TGF-β-signaling in tumor progression, we propose HF as a potential new anti-cancer drug

Acknowledgements

Halofuginone was obtained as a kind donation from Prof. Arnon Nagler, Chaim Sheba Medical Center, Tel Hashomer, Israel. The transgenic hCG-PML-RARα mice were kindly provided by Prof. Pier Paolo Pandolfi (Beth Israel Deaconess Medical Center, Harvard Stem Cell Institute, Boston, MA, USA). This work was supported by Fundação de Amparo à Pesquisa do Estado de São Paulo (FAPESP, grant No. 2013/08135-2). P.A.A. was an FAPESP fellowship recipient (Grant No. 08/52634-5).
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.
The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.
The Creative Commons Public Domain Dedication waiver (https://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

§PAA and LLFP have equally contributed to the study. PAA, LLFP, NA and EMR: Conceived and designed the experiments; PAA, LLFP, ASGL, VL, LAC, CTP, ASGL and FPS: Performed the experiments; PAA, LLFP, VL, FPS and FC: Analyzed the data; RAP, FC, AN, RPF and EMR: Contributed reagents/materials/analysis tools; PAA and EMR: Wrote the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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
7.
8.
Zurück zum Zitat 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.
Zurück zum Zitat Ayala F, Dewar R, Kieran M, Kalluri R. Contribution of bone microenvironment to leukemogenesis and leukemia progression. Leukemia. 2009;23:2233–41.PubMedCentralPubMedCrossRef Ayala F, Dewar R, Kieran M, Kalluri R. Contribution of bone microenvironment to leukemogenesis and leukemia progression. Leukemia. 2009;23:2233–41.PubMedCentralPubMedCrossRef
10.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat Roberts AB. TGF-beta signaling from receptors to the nucleus. Microbes Infect. 1999;1:1265–73.PubMedCrossRef Roberts AB. TGF-beta signaling from receptors to the nucleus. Microbes Infect. 1999;1:1265–73.PubMedCrossRef
40.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 2003;3:401–10.PubMedCrossRef Bergers G, Benjamin LE. Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 2003;3:401–10.PubMedCrossRef
45.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat You WK, McDonald DM. The hepatocyte growth factor/c-Met signaling pathway as a therapeutic target to inhibit angiogenesis. BMB Rep. 2008;41:833–9.PubMedCentralPubMedCrossRef You WK, McDonald DM. The hepatocyte growth factor/c-Met signaling pathway as a therapeutic target to inhibit angiogenesis. BMB Rep. 2008;41:833–9.PubMedCentralPubMedCrossRef
49.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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.
Zurück zum Zitat 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
Metadaten
Titel
Halofuginone inhibits phosphorylation of SMAD-2 reducing angiogenesis and leukemia burden in an acute promyelocytic leukemia mouse model
verfasst von
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
Publikationsdatum
01.12.2015
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2015
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-015-0181-2

Weitere Artikel der Ausgabe 1/2015

Journal of Experimental & Clinical Cancer Research 1/2015 Zur Ausgabe

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Labor, CT-Anthropometrie zeigen Risiko für Pankreaskrebs

13.05.2024 Pankreaskarzinom Nachrichten

Gerade bei aggressiven Malignomen wie dem duktalen Adenokarzinom des Pankreas könnte Früherkennung die Therapiechancen verbessern. Noch jedoch klafft hier eine Lücke. Ein Studienteam hat einen Weg gesucht, sie zu schließen.

Viel pflanzliche Nahrung, seltener Prostata-Ca.-Progression

12.05.2024 Prostatakarzinom Nachrichten

Ein hoher Anteil pflanzlicher Nahrung trägt möglicherweise dazu bei, das Progressionsrisiko von Männern mit Prostatakarzinomen zu senken. In einer US-Studie war das Risiko bei ausgeprägter pflanzlicher Ernährung in etwa halbiert.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.