The growth of new lymphatic vessels (lymphangiogenesis) in tumors is an integral step in the metastatic spread of tumor cells, first to the sentinel lymph nodes that surround the tumor and then elsewhere in the body. Currently, no selective agents designed to prevent lymphatic vessel growth have been approved for clinical use, and there is an important potential clinical niche for antilymphangiogenic agents. Using a zebrafish phenotype-based chemical screen, we have identified drug compounds, previously approved for human use, that have antilymphatic activity. These include kaempferol, a natural product found in plants; leflunomide, an inhibitor of pyrimidine biosynthesis; and cinnarizine and flunarizine, members of the type IV class of calcium channel antagonists. Antilymphatic activity was confirmed in a murine in vivo lymphangiogenesis Matrigel plug assay, in which kaempferol, leflunomide, and flunarizine prevented lymphatic growth. We show that kaempferol is a novel inhibitor of VEGFR2/3 kinase activity and is able to reduce the density of tumor-associated lymphatic vessels as well as the incidence of lymph node metastases in a metastatic breast cancer xenograft model. However, in this model, kaempferol administration was also associated with tumor deposits in the pancreas and diaphragm, and flunarizine was found to be tumorigenic. Although this screen revealed that zebrafish is a viable platform for the identification and development of mammalian antilymphatic compounds, it also highlights the need for focused secondary screens to ensure appropriate efficacy of hits in a tumor context. Mol Cancer Ther; 13(10); 2450–62. ©2014 AACR.

This article is featured in Highlights of This Issue, p. 2251

Cancer is now the leading cause of death worldwide. The majority of cancer-induced deaths are caused by metastatic malignancy, a process by which cancer cells spread through the body via the lymphatic or blood vasculature. The importance of lymphatic metastases is well recognized in cancer staging and treatment, with the spread of cancer cells to surrounding lymph nodes being associated with poor prognosis in many cancers (1, 2). It has been estimated that more than 80% of solid tumors metastasize, at least partially, through the lymphatic vasculature (3). Tumor cells can enter the lymphatic vasculature by either invading preexisting lymphatic vessels present in the tissue surrounding the tumor, or by promoting lymphangiogenesis and creating new lymphatic vessels within and around the tumor—a process termed tumor-induced lymphangiogenesis (4, 5). The increase in lymphatic vessel density in the tumor has been proposed to facilitate the metastatic spread of cancer cells, as it has been correlated with an increased incidence of lymph node metastases and a consequential decrease in patient survival in many cancers (3, 6). As well as being implicated in cancer metastasis, de novo lymphangiogenesis is also associated with host rejection of renal transplants (7) and corneal grafts (8).

Studies have shown that the receptor tyrosine kinases VEGFR2 and VEGFR3 as well as their ligands VEGF-A, VEGF-C, and VEGF-D play important roles in tumor-induced lymphangiogenesis (4, 9–12). It is known that inhibition of the VEGFR signaling pathway reduces tumor-associated lymphangiogenesis and lymph node metastasis in animal models (13–16) and also improves the survival of corneal transplants (8). Therefore, there is a need to develop and test not only inhibitors of VEGFR/VEGF signaling but also to identify novel antilymphatic drugs that act outside of this pathway.

Zebrafish are a powerful model organism in which to study lymphangiogenesis. Lymphatic vessels can be readily observed in transparent embryos, and zebrafish lymphatic development shares the same molecular mechanisms as used by mammals (17–19). Zebrafish embryos, like cell-based assays, are also amenable to small-molecule drug screens (20). In zebrafish, the mechanism of lymphatic vessel development in the trunk has been well established, making it ideal for antilymphatic studies. At 36 hours postfertilization (hpf), lymphatic precursors known as secondary sprouts migrate from the posterior cardinal vein (PCV) and by 48 hpf have migrated to the horizontal myoseptum where they are termed parachordal lymphangioblasts. From here, the lymphangioblasts migrate toward and then along the arterial intersegmental blood vessels (ISV) to form the thoracic duct by 120 hpf (21). Although small-molecule drug screens using zebrafish embryos have been used to successfully identify inhibitors of mammalian blood vessel development (22–24), no antilymphatic screens have been reported to date.

One prerequisite for conducting an antilymphatic chemical screen is a specific marker for lymphatic vessels. We recently identified and characterized the expression of the zebrafish ortholog of lymphatic vessel endothelial hyaluronan receptor 1 (lyve1; ref. 25). LYVE-1 is extensively used as a marker for lymphatic vessels in mice and humans, and we found zebrafish lyve1 to be an excellent marker for lymphatic development as it is expressed in the veins and the developing lymphatic vessels (26). We have also shown that treatment of zebrafish embryos with known lymphatic inhibitors reduced or abolished the expression of lyve1 mRNA (25).

In this study, we utilized the zebrafish model to identify novel inhibitors of mammalian lymphangiogenesis. We undertook a chemical screen in which compounds were initially identified by the ability to reduce lyve1 expression in zebrafish embryos, before being passed through secondary screens to confirm that they specifically inhibited lymphatic vessel development in zebrafish. Finally, lead compounds were characterized in a mammalian model of lymphangiogenesis and their efficacy in preventing lymph node metastases was determined in a metastatic breast cancer xenograft model.

Zebrafish

The following lines were used in this study: wild-type (AB), TG(lyve1:egfp)nz150 (26), TG(fli1:egfp)y1 (27), and TG(kdrl:nlsmCherry)is4 (28).

Compounds

The Prestwick Chemical Library was supplied at 2 mg/mL in DMSO. Compounds were diluted to 5 μg/mL in screening media consisting of 1 mmol/L Tris (pH 7.5) and 1x penicillin/streptomycin (Gibco) made up in E3 media. Additional compounds were sourced as follows: leflunomide (CAS #75706-12-6), cinnarizine (CAS #298-57-7), and flunarizine dihydrochloride (CAS #30484-77-6) from Sigma, A77 1726 (CAS #108605-62-5) from Calbiochem, kaempferol (CAS #520-18-3) from Ivy Fine Chemicals, and rapamycin (CAS #53123-88-9) from Life Research and Selleck Chemicals. For animal studies, the following vehicles were used: 2.5% ethanol (Merck), 5% polyethylene glycol 400 (Sigma), and 5% Tween-80 (Sigma) for rapamycin; 20% 2-hydroxypropyl-β-cyclodextrin (Sigma) for flunarizine; and 1% and 1.5% carboxymethylcellulose (Sigma) for kaempferol and leflunomide, respectively.

Whole-mount in situ hybridization

In situ hybridization was performed using a lyve1 antisense probe (25) in the Biolane HTI in situ robot (Hölle and Hüttner), as described previously (29).

Zebrafish image analysis and statistical analysis

Embryos were imaged as described (30). Thoracic duct formation at 120 hpf and secondary sprout formation at 36 hpf were scored as described previously (26). Lymphangioblast cell tracking for the first 8 hours following their emergence from the PCV was performed manually using ImageJ (NIH, Bethesda, MD). Statistical analysis was performed using Prism 5.0 software (GraphPad Software Inc). Significance was determined by Mann–Whitney tests.

Mouse studies

All mouse experiments followed protocols approved by the Animal Ethics Committee of the University of Auckland (Auckland, New Zealand). Age-matched female C57BL/6 mice and NIH-III nude mice (NIH-LystbgFoxn1nuBtkxid) weighing between 18 g and 25 g were provided by the Vernon Jansen Unit, University of Auckland.

Matrigel plug assay

Female C57BL/6 mice were anesthetized with ketamine (100 mg/kg; Parnell Living Science) and xylazine (10 mg/kg; Phoenix Pharmaceuticals Inc), followed by shaving of the left flank and subcutaneous injection of 500 μL Matrigel (Becton Dickinson) supplemented with VEGF-C (1 μg/mL; R&D Systems). Mice were randomly assigned to treatment groups dosed at maximum tolerated dose (MTD; 200 mg/kg kaempferol, 200 mg/kg leflunomide, and 30 mg/kg flunarizine; n = 4 mice/group) 1 hour post Matrigel implantation, and then daily for a further 9 days. Plugs were then harvested and frozen in liquid nitrogen for cryostat sectioning into 5-μm fresh-frozen sections.

Cell culture

MDA-MB-231-luc D3H2LN cells were supplied by Caliper Life Sciences (November 2009) and authenticated by short tandem repeat profiling at CellBank Australia in September 2011. Cells were confirmed to be Mycoplasma-negative by PCR testing (Boehringer Mannheim) and were passaged in culture for less than 6 months after authentication. Cells were cultured in α-minimum essential media containing 10% fetal calf serum, 100 U/mL penicillin, and 100 μg/mL streptomycin, and incubated at 37°C in a 5% CO2 humidified incubator. Cells were used in experiments during the exponential phase of growth (60%–80% confluency).

Orthotopic xenograft study

The fourth mammary fat pads of NIH III nude mice were inoculated with 3 × 106 MDA-MB-231-luc D3H2LN cells suspended in Matrigel and PBS. At 48 hours, mice with detectable tumor cells were recruited into the prevention regimen, in which daily dosing began (control; n = 4 mice/group; 150 mg/kg kaempferol, 40 mg/kg flunarizine, and 5 mg/kg rapamycin; n = 6 mice/group). The remaining mice were left untreated until week 4 when mice with established tumors were recruited into the intervention regimen and daily dosing began (control; n = 4 mice/group; 150 mg/kg kaempferol and 40 mg/kg flunarizine; n = 6 mice/group). Control prevention and intervention regimens were pooled for analysis. Primary tumor growth was measured every 2 to 3 days with digital calipers. At 7 to 8 weeks posttumor inoculation, mice were euthanized and their primary tumors, selected lymph nodes (brachial, axillary, and inguinal), pancreases, diaphragms, and any other tissues with visible metastases were harvested. Tissues were then fixed in 10% neutral-buffered formalin, paraffin-embedded, and 5 μm fixed tissue sections prepared for IHC.

Bioluminescence imaging

Mice were anesthetized with isoflurane (Lunan Pharmaceutical Corporation) and then imaged with the IVIS Kinetic imager (Caliper Life Sciences) approximately 10 minutes after subcutaneous administration of 150 mg/kg D-Luciferin firefly potassium salt (Gold Biotechnology) in PBS. Imaging was carried out 24 hours following tumor cell inoculation to confirm the success of the procedure, and then weekly from week 3. After week 4, mice were probed for secondary metastatic signals by shielding the primary tumor with black paper and taping back the forelimbs to expose the brachial/axillary lymph node areas. Tissues harvested postmortem were imaged ex vivo in 24-well tissue culture plates containing 300 μg/mL luciferin.

Immunohistochemistry

For both tumor and Matrigel sections, lymphatic vessels were visualized by IHC using primary rabbit anti-mouse LYVE1 (1:150; Angiobio) and rat anti-mouse F4/80 (1:50; Invitrogen) antibodies, and secondary goat anti-rat AlexaFluor 488 and goat anti-rabbit AlexaFluor 568 antibodies (1:200; Invitrogen), followed by counterstaining with DAPI (Invitrogen). Imaging of sections was carried out using a Zeiss Axio Imager Z2 microscope for whole section scanning and an Olympus FV1000 confocal microscope for high-resolution imaging. Lymphatic vessel density was calculated as the number of LYVE1+/F480 vessels per μm2 of either tumor or Matrigel plug and graphed as a percentage of control. A minimum of two sections covering the entire area of the tumor or Matrigel plug was used per animal. Statistical analysis was performed using Prism 6.0 software (GraphPad Software Inc), and significance was determined by one-way ANOVA.

A chemical screen in zebrafish identified drugs with antilymphatic activity

The Prestwick Chemical Library, containing 1,120 compounds (the majority of which are FDA-approved drugs) was selected for our screen. Using our antilymphatic screening assay (see Supplementary Methods), we identified 12 compounds, a hit rate of 1.1%, which reduced or abolished lyve1 expression without adverse side-effects such as developmental delay or tissue necrosis. The mTOR inhibitor, rapamycin, was included as a positive control as it is a known inhibitor of both mammalian and zebrafish lymphangiogenesis (25, 31, 32). A secondary screen for thoracic duct formation using lyve1:egfp embryos (26) showed that six compounds displayed antilymphatic activity in zebrafish: the flavonoid kaempferol, the dihydroorotate dehydrogenase inhibitor leflunomide, 2 diphenylpiperazine calcium channel antagonists, flunarizine and cinnarizine and 2 statins, lovastastin, and simvastatin (Fig. 1A and Supplementary Fig. S1). The identification of statins as antilymphatic was expected as both compounds were recently shown to have antilymphatic properties (33). The secondary screen using fli1:egfp embryos identified kaempferol, leflunomide, cinnarizine, and flunarizine as agents that prevented lymphangiogenesis in the zebrafish without disrupting blood vessel development, whereas lovastatin and simvastatin disrupted ISV development at the screening dose of 5 μg/mL. When the concentration of each statin was reduced so that they no longer blocked ISV formation, the thoracic duct formed normally (Supplementary Fig. S2). As lovastatin and simvastatin are not specific inhibitors of lymphangiogenesis, we did not consider these compounds for further analysis.

Figure 1.

Identification of compounds that inhibit lymphatic vessel development. A, representative images of the positive hits from the lyve1 in situ hybridization screen of the Prestwick compound library. Of note, 500 nmol/L rapamycin was included as a positive control. B, confocal images of 120 hpf lyve1:egfp embryos treated with potential antilymphatic drugs from 24 hpf. Embryos treated with 1% DMSO have normal lymphatic development with the thoracic duct (arrows) forming, whereas embryos treated with either kaempferol, leflunomide, A77 1726, cinnarizine, flunarizine, or rapamycin have no thoracic duct. C, quantitation of thoracic duct formation at 120 hpf in embryos treated with antilymphatic compounds from 24 hpf. Error bars ± SD. Scale bar, 50 μm.

Figure 1.

Identification of compounds that inhibit lymphatic vessel development. A, representative images of the positive hits from the lyve1 in situ hybridization screen of the Prestwick compound library. Of note, 500 nmol/L rapamycin was included as a positive control. B, confocal images of 120 hpf lyve1:egfp embryos treated with potential antilymphatic drugs from 24 hpf. Embryos treated with 1% DMSO have normal lymphatic development with the thoracic duct (arrows) forming, whereas embryos treated with either kaempferol, leflunomide, A77 1726, cinnarizine, flunarizine, or rapamycin have no thoracic duct. C, quantitation of thoracic duct formation at 120 hpf in embryos treated with antilymphatic compounds from 24 hpf. Error bars ± SD. Scale bar, 50 μm.

Close modal

Kaempferol, leflunomide, cinnarizine, and flunarizine reduced lyve1 expression in our in situ screen and prevented thoracic duct formation in either fli1:egfp or lyve1:egfp embryos (Fig. 1B and C and Supplementary Fig. S1). Using thoracic duct formation as a readout of drug efficacy, we established a response curve and MTD for each compound in zebrafish embryos (Fig. 1C). The MTD for kaempferol was 30 μmol/L, leflunomide 4 μmol/L, cinnarizine 28 μmol/L, and flunarizine 5 μmol/L. These doses were used for all subsequent zebrafish experiments. We also tested the active metabolite of leflunomide, A77 1726 (34, 35), and found that it also inhibited lymphatic vessel development with an MTD of 2 μmol/L.

Kaempferol and leflunomide inhibit the sprouting of zebrafish lymphatic vessels

To establish which aspect of lymphatic vessel development each drug was inhibiting, we imaged the trunk region of lyve1:egfp embryos at 36 hpf and 48 hpf. We scored the number of secondary sprouts at 36 hpf and found that embryos treated with either kaempferol, leflunomide, or A77 1726 had reduced numbers of secondary sprouts, whereas cinnarizine or flunarizine-treated embryos were normal (Fig. 2A and B). We also scored the number of parachordal lymphangioblasts at 48 hpf and found that kaempferol, leflunomide, and A77 1726 caused a reduction in lymphangioblasts, consistent with a defect in secondary sprouting. However, both cinnarizine and flunarizine-treated embryos had normal lymphangioblast formation (Fig. 2C and D).

Figure 2.

Kaempferol and leflunomide inhibit secondary sprouting. A, confocal images of 36 hpf lyve1:egfp embryos treated with either 1% DMSO, kaempferol, leflunomide, A77 1726, cinnarizine, or flunarizine. Secondary sprouts from the PCV (arrowheads) are not present in embryos treated with kaempferol, leflunomide, or A77 1726. B, quantitation of secondary sprout formation at 36 hpf in embryos treated with antilymphatic compounds. C, confocal projection images of 48 hpf lyve1:egfp embryos treated with either 1% DMSO, kaempferol, leflunomide, A77 1726, cinnarizine, or flunarizine. Parachordal lymphangioblasts (arrows) are not present in embryos treated with kaempferol, leflunomide, or A77 1726. D, percentage of somites containing parachordal lymphangioblasts at 48 hpf in embryos treated with antilymphatic compounds. Error bars ± SD. ***, P < 0.001; **, P < 0.01, ns > 0.05 by a Mann–Whitney test to 1% DMSO control. Scale bar, 50 μm.

Figure 2.

Kaempferol and leflunomide inhibit secondary sprouting. A, confocal images of 36 hpf lyve1:egfp embryos treated with either 1% DMSO, kaempferol, leflunomide, A77 1726, cinnarizine, or flunarizine. Secondary sprouts from the PCV (arrowheads) are not present in embryos treated with kaempferol, leflunomide, or A77 1726. B, quantitation of secondary sprout formation at 36 hpf in embryos treated with antilymphatic compounds. C, confocal projection images of 48 hpf lyve1:egfp embryos treated with either 1% DMSO, kaempferol, leflunomide, A77 1726, cinnarizine, or flunarizine. Parachordal lymphangioblasts (arrows) are not present in embryos treated with kaempferol, leflunomide, or A77 1726. D, percentage of somites containing parachordal lymphangioblasts at 48 hpf in embryos treated with antilymphatic compounds. Error bars ± SD. ***, P < 0.001; **, P < 0.01, ns > 0.05 by a Mann–Whitney test to 1% DMSO control. Scale bar, 50 μm.

Close modal

Confirmation of these phenotypes was obtained by time-lapse imaging of lyve1:egfp embryos, in the absence (Supplementary Video S1) and presence of the antilymphatic compounds, from 30 to 49 hpf. In this way, we confirmed that kaempferol causes a defect in secondary sprouting, as no secondary sprouts were observed in three time-lapse experiments (Fig. 3; Supplementary Video S2). Leflunomide caused secondary sprouts to migrate at half the speed observed in control embryos (4.9 μm/hour in leflunomide compared with 11.4 μm/hour in control) and each sprout had an abnormal morphology, with multiple vascular tips (Fig. 3; Supplementary Video S3). The secondary sprouts in embryos treated with flunarizine looked normal and migrated at comparable speed to controls (Fig. 3; Supplementary Video S4). Our results suggest that kaempferol and leflunomide inhibit the process of lymphatic sprouting from the veins and the type IV calcium channel agonists, cinnarizine and flunarizine, inhibit later stages of lymphatic vessel formation.

Figure 3.

Live-imaging lymphangioblast formation in the presence of antilymphatic compounds. A, stills from time-lapse imaging of lyve1:egfp embryos from 30 hpf to 49:42 hpf (19:42 hours) treated with either 1% DMSO (Supplementary Video S1), kaempferol (Supplementary Video S2), leflunomide (Supplementary Video S3), or flunarizine (Supplementary Video S4) from 24 hpf. Parachordal lymphangioblasts (arrows) do not form in embryos treated with kaempferol or leflunomide. B, cell tracks of lymphangioblasts (red lines) over 8 hours as they migrate from the PCV (black line), with the average cell speed given in μm per hour. At least eight lymphangioblasts were tracked per treatment and data are collated from three independent time-lapse experiments. Scale bar, 20 μm.

Figure 3.

Live-imaging lymphangioblast formation in the presence of antilymphatic compounds. A, stills from time-lapse imaging of lyve1:egfp embryos from 30 hpf to 49:42 hpf (19:42 hours) treated with either 1% DMSO (Supplementary Video S1), kaempferol (Supplementary Video S2), leflunomide (Supplementary Video S3), or flunarizine (Supplementary Video S4) from 24 hpf. Parachordal lymphangioblasts (arrows) do not form in embryos treated with kaempferol or leflunomide. B, cell tracks of lymphangioblasts (red lines) over 8 hours as they migrate from the PCV (black line), with the average cell speed given in μm per hour. At least eight lymphangioblasts were tracked per treatment and data are collated from three independent time-lapse experiments. Scale bar, 20 μm.

Close modal

Kaempferol inhibits VEGFR kinase activity

The IC50 values for kaempferol, leflunomide, A77 1726, and flunarizine were determined against human VEGFR1, VEGFR2, and VEGFR3. Only kaempferol caused appreciable inhibition of VEGFR kinase activity with an IC50 of 8.37 μmol/L against VEGFR2 and 19 μmol/L against VEGFR3 (Supplementary Table S1). These data suggest that the antilymphatic activity we observe following kaempferol treatment is most likely through the inhibition of VEGFR2/3 signaling and that leflunomide and flunarizine function through other pathways.

Flunarizine causes lymphangioblast cell death in zebrafish

To determine the mechanism by which the type IV calcium channel inhibitors prevent lymphatic vessel formation, we imaged lyve1:egfp;kdrl:nls:mcherry embryos exposed to flunarizine at later stages of lymphatic development, from 48 hpf until 68 hpf. We took advantage of residual kdrl expression in parachordal lymphangioblasts and used a nuclear-localized kdrl transgenic, kdrl:nls:mcherry, to fate-map lymphangioblasts exposed to flunarizine. By tracking the nuclei of lymphangioblasts (lyve1 and kdrl positive) throughout three independent time-lapse experiments, we observed that in embryos exposed to flunarizine, on average 37% of their lymphangioblasts underwent nuclear fragmentation and then subsequently lost lyve1 and kdrl expression, indicative of cell death by apoptosis. In contrast, we never observed any lymphangioblast cell death in control embryos (Fig. 4; Supplementary Videos S5 and S6). Next, we performed terminal deoxynucleotidyl transferase–mediated dUTP nick end labeling (TUNEL) to identify cells undergoing apoptosis at 60 hpf in both control and flunarizine-treated embryos. We found an average of 2.0 TUNEL-positive lymphangioblasts per embryo with flunarizine compared with 0.1 in control embryos (Supplementary Fig. S3). Our data suggest that flunarizine, while not affecting initial specification and migration of lymphangioblasts, causes them to undergo apoptosis, leading to defects in lymphatic vessel development.

Figure 4.

Lymphangioblasts do not survive in embryos treated with flunarizine. Stills from time-lapse imaging of lyve1:egfp;kdrl:nls:mCherry embryos from 48 hpf (A and K) to 68 hpf (B and L) treated with either 1% DMSO (A–J, Supplementary Video S5) or 5 μmol/L flunarizine (K–T, Supplementary Video S6) from 24 hpf. kdrl and lyve1-positive cells numbered 1 and 2 in A and K. C–J and M–T are cropped images of the kdrl:nls:mCherry channel (dotted line in A, B, K, L) with the numbered lymphangioblasts tracked at different time points. The two tracked lymphangioblasts survive in the embryo treated with 1% DMSO. C–J, in the embryo treated with flunarizine, both the #1 and #2.1 lymphangioblasts lose kdrl and lyve1 expression over the time-lapse (M–T) with their nuclei appearing to fragment. U, quantitation of the percentage of parachordal lymphangioblasts that die per time-lapse experiment (from 48 hpf to 68 hpf) in embryos treated with 1% DMSO or flunarizine. Error bars ± SD. *, P < 0.05 by a Mann–Whitney test to 1% DMSO control. Scale bar, 20 μm.

Figure 4.

Lymphangioblasts do not survive in embryos treated with flunarizine. Stills from time-lapse imaging of lyve1:egfp;kdrl:nls:mCherry embryos from 48 hpf (A and K) to 68 hpf (B and L) treated with either 1% DMSO (A–J, Supplementary Video S5) or 5 μmol/L flunarizine (K–T, Supplementary Video S6) from 24 hpf. kdrl and lyve1-positive cells numbered 1 and 2 in A and K. C–J and M–T are cropped images of the kdrl:nls:mCherry channel (dotted line in A, B, K, L) with the numbered lymphangioblasts tracked at different time points. The two tracked lymphangioblasts survive in the embryo treated with 1% DMSO. C–J, in the embryo treated with flunarizine, both the #1 and #2.1 lymphangioblasts lose kdrl and lyve1 expression over the time-lapse (M–T) with their nuclei appearing to fragment. U, quantitation of the percentage of parachordal lymphangioblasts that die per time-lapse experiment (from 48 hpf to 68 hpf) in embryos treated with 1% DMSO or flunarizine. Error bars ± SD. *, P < 0.05 by a Mann–Whitney test to 1% DMSO control. Scale bar, 20 μm.

Close modal

Antilymphatic drugs identified in zebrafish inhibit mammalian lymphangiogenesis

To confirm that the antilymphatic compounds identified could also inhibit mammalian lymphangiogenesis, they were administered to C57BL/6 mice injected with Matrigel plugs supplemented with recombinant VEGF-C. The amount of lymphatic vessel growth into the plug was determined by immunostaining with anti-LYVE1. To rule out any confounding LYVE1 staining in macrophages (36), we also performed immunostaining with an F4/80 antibody against macrophage cells and selected LYVE1-positive F4/80-negative cells as being lymphatic. As expected, rapamycin was able to significantly reduce the number of lyve1 vessels when compared with controls. Of note, 200 mg/kg kaempferol, 200 mg/kg leflunomide, and 30 mg/kg flunarizine all significantly reduced the infiltration of new lymphatic vessels relative to controls (Fig. 5). On the basis of these results, kaempferol and flunarizine were selected for efficacy testing in an orthotopic breast cancer xenograft study.

Figure 5.

Kaempferol and flunarizine inhibit mammalian lymphangiogenesis. A, confocal images from immunofluorescent sections of Matrigel plugs costained with anti-LYVE-1 (red) and anti-F4/80 (green). Lymphatic vessels are LYVE-1 positive, F4/80 negative (arrows). B, quantitation of lymphatic vessel density in Matrigel plugs. Error bars ± SD. *, P < 0.05 by one-way ANOVA; **, P < 0.01 by one-way ANOVA. Scale bar, 100 μm.

Figure 5.

Kaempferol and flunarizine inhibit mammalian lymphangiogenesis. A, confocal images from immunofluorescent sections of Matrigel plugs costained with anti-LYVE-1 (red) and anti-F4/80 (green). Lymphatic vessels are LYVE-1 positive, F4/80 negative (arrows). B, quantitation of lymphatic vessel density in Matrigel plugs. Error bars ± SD. *, P < 0.05 by one-way ANOVA; **, P < 0.01 by one-way ANOVA. Scale bar, 100 μm.

Close modal

Kaempferol reduces lymph node metastases but increases systemic metastases in orthotopic breast cancer xenografts

To test whether the antilymphatic activity of kaempferol and flunarizine could reduce lymphatic metastasis, we used an orthotopic breast cancer xenograft model in which metastatic human breast cancer cells expressing luciferase (MDA-MB-231-luc-D3H2LN) were injected into the mammary fat pad of NIH-III nude mice. Animals were treated with drugs by two different regimens: prevention treatment, in which mice were dosed daily with antilymphatic drugs 48 hours posttumor implantation and intervention treatment, which was initiated 28 days posttumor implantation, just before the onset of metastases detection in controls. The dose of flunarizine was increased to 40 mg/kg to attempt to enhance efficacy, whereas kaempferol was reduced to 150 mg/kg to improve tolerance. Both drugs, and the control drug rapamycin, were well tolerated and did not cause any adverse body weight change in the NIH-III nude mice (Supplementary Fig. S4). Rapamycin inhibited primary tumor growth, whereas kaempferol had minimal effect on tumor volume and flunarizine promoted tumor growth in both prevention and intervention groups as determined by caliper measurement and bioluminescence imaging. (Fig. 6A and B and Supplementary Fig. S5). Primary tumors were removed at the conclusion of treatment, 7 to 8 weeks after tumor cell inoculation, to assess the density of tumor-associated lymphatics (which includes both peritumoral and intratumoral lymphatic vessels) by LYVE1-positive F4/80-negative staining. Kaempferol was effective at reducing the density of tumor-associated lymphatics in both the prevention and intervention groups and any lymphatics we observed were often smaller and appeared collapsed compared with controls. In contrast, tumors from flunarizine-treated mice had similar levels of tumor-associated lymphatics to control tumors (Fig. 6C and D).

Figure 6.

The effect of drugs on tumor growth and tumor lymphangiogenesis. A and B, tumor volume in NIH-III nude mice, orthotopically xenografted with MDA-MB-231-luc-D3H2LN cells, in which daily drug dosing began (A) 48 hours posttumor implantation (prevention group) or (B) 28 days posttumor implantation (intervention group). Error bars, ± SEM. C, images of tumor sections costained with anti-LYVE-1 (red) and anti-F4/80 (green). Lymphatic vessels are LYVE-1-positive, F4/80-negative (arrows). The tumor (T) boundary is marked with a dotted line. D, quantitation of intratumoral and peritumoral lymphatic vessel density in tumors. Error bars ± SD. *, P < 0.05; **, P < 0.01 by one-way ANOVA. Scale bar, 100 μm.

Figure 6.

The effect of drugs on tumor growth and tumor lymphangiogenesis. A and B, tumor volume in NIH-III nude mice, orthotopically xenografted with MDA-MB-231-luc-D3H2LN cells, in which daily drug dosing began (A) 48 hours posttumor implantation (prevention group) or (B) 28 days posttumor implantation (intervention group). Error bars, ± SEM. C, images of tumor sections costained with anti-LYVE-1 (red) and anti-F4/80 (green). Lymphatic vessels are LYVE-1-positive, F4/80-negative (arrows). The tumor (T) boundary is marked with a dotted line. D, quantitation of intratumoral and peritumoral lymphatic vessel density in tumors. Error bars ± SD. *, P < 0.05; **, P < 0.01 by one-way ANOVA. Scale bar, 100 μm.

Close modal

In addition to the primary tumor, the brachial, axillary, and inguinal lymph nodes surrounding the tumors as well as the pancreas and diaphragm were removed and were imaged ex vivo for any luciferase activity that would indicate that cancer cells had metastasized (Fig. 7A). The frequency of lymph nodes with a positive bioluminescent signal was significantly reduced in rapamycin and kaempferol prevention groups relative to controls; however, flunarizine-treated mice had a significant increase in the number of lymph nodes with a bioluminescent signal. In the intervention arm, kaempferol was similarly effective, reducing the frequency of lymph nodes with a positive bioluminescent signal (Fig. 7B).

Figure 7.

Kaempferol reduces metastasis to lymph nodes but increases the frequency of systemic metastasis. A, in vivo and ex vivo bioluminescent imaging of NIH-III nude mice, orthotopically xenografted with MDA-MB-231-luc-D3H2LN human breast cancer cells. B and C, quantitation of the metastatic spread of tumor cells as determined by the presence of bioluminescence in either lymph nodes (B) or in pancreas and diaphragm (systemic tissues, C). D–I, H&E staining of tissue confirming the presence of neoplastic cells in the lymph nodes of control (D) and flunarizine (F) treated mice but not in kaempferol mice (E). Neoplastic cells shown within dotted line, arrowheads show mitotic figures. D′–F′ are magnified images of D–F. H&E staining of the pancreas in control (G) and in kaempferol-treated mice (H; arrows indicate normal tissue surrounded by neoplastic cells) and in the diaphragm of flunarizine mice (I; neoplastic cells shown within dotted line). Error bars ± SEM. *, P < 0.05; **, P < 0.01 by one-way ANOVA. Scale bars, 100 μm (F and I) and 25 μm (F′).

Figure 7.

Kaempferol reduces metastasis to lymph nodes but increases the frequency of systemic metastasis. A, in vivo and ex vivo bioluminescent imaging of NIH-III nude mice, orthotopically xenografted with MDA-MB-231-luc-D3H2LN human breast cancer cells. B and C, quantitation of the metastatic spread of tumor cells as determined by the presence of bioluminescence in either lymph nodes (B) or in pancreas and diaphragm (systemic tissues, C). D–I, H&E staining of tissue confirming the presence of neoplastic cells in the lymph nodes of control (D) and flunarizine (F) treated mice but not in kaempferol mice (E). Neoplastic cells shown within dotted line, arrowheads show mitotic figures. D′–F′ are magnified images of D–F. H&E staining of the pancreas in control (G) and in kaempferol-treated mice (H; arrows indicate normal tissue surrounded by neoplastic cells) and in the diaphragm of flunarizine mice (I; neoplastic cells shown within dotted line). Error bars ± SEM. *, P < 0.05; **, P < 0.01 by one-way ANOVA. Scale bars, 100 μm (F and I) and 25 μm (F′).

Close modal

Although kaempferol was effective at reducing lymph node metastases, 80% of mice treated with kaempferol had MDA-MB-231-luc-D3H2LN cells present in the pancreas and diaphragm in both the prevention and intervention regimens, compared with 10% of control mice as determined by bioluminescence imaging. No evidence of bioluminescence was observed in pancreas or diaphragm tissue in rapamycin-treated mice in the prevention regimen (Fig. 7C) or in flunarizine intervention mice, but was in 50% of flunarizine prevention mice. The presence of cancer cell infiltration into lymph node, pancreas, and diaphragm tissue was confirmed by histopathological evaluation of hematoxylin and eosin (H&E)-stained sections (Fig. 7D–I).

In this study, we identified four compounds that prevented lymphangiogenesis in zebrafish. The first compound, kaempferol, a natural flavonoid that can be isolated from many plant sources such as broccoli, apples, and tea (37), was able to inhibit secondary sprouting from the PCV in zebrafish embryos. This phenotype is identical to the phenotype observed when the Vegfr3/Vegfc signaling pathway is impaired (17, 19), and in support of this observation, we find that kaempferol is able to inhibit human VEGFR2 and VEGFR3 at low micromolar concentrations. We also found that kaempferol had good efficacy at preventing mammalian lymphangiogenesis into Matrigel plugs and at reducing the density of tumor-associated lymphatics, as well as the frequency of lymph node metastases when dosed either as a prevention or intervention drug. Although lymph node metastases were reduced with kaempferol treatment, we observed an increase in the incidence of metastases in the pancreas and diaphragm. It is unclear whether the increase in systemic metastases results from a compensatory mechanism to overcome the inhibition of tumor lymphatic growth, or if kaempferol is somehow promoting nonlymphatic spread of cancer cells. It is also uncertain whether these metastases are the result of hematogenic spread or the local invasion of tumor cells. Regardless, these results suggest that antilymphatics may be best given in conjunction with other compounds that prevent tumor growth. Previous work has shown that kaempferol consumption has been associated with reduced risk of developing many cancers (38–40) and has also been shown to reduce VEGF expression in human cancer cell lines (41, 42). To our knowledge, this study is the first to report VEGFR inhibition and antilymphatic activity for kaempferol.

Our second antilymphatic compound, leflunomide, is an inhibitor of the enzyme dihydroorotate dehydrogenase (DHODH), involved in pyrimidine biosynthesis (43, 44). Leflunomide is used as a disease-modifying drug to treat patients with rheumatoid arthritis (45) and has also been shown to have antitumor properties in models of melanoma (46). In this study, we found that leflunomide and its active metabolite, A77 1726, are able to inhibit secondary sprouting of lymphatic precursors in zebrafish embryos and it is also able to inhibit lymphangiogenesis in a murine Matrigel plug assay. We believe that leflunomide is likely to be acting as an antilymphatic drug through its ability to inhibit DHODH as the low micromolar doses we used in zebrafish are unable to inhibit human VEGFR signaling and are consistent with leflunomide functioning as an inhibitor of pyrimidine biosynthesis (35, 46). It is possible that as lymphangioblasts migrate and remodel to form vessels, they are also sensitive to reduced levels of pyrimidines and therefore by inhibiting de novo pyrimidine biosynthesis in the lymphangioblasts, leflunomide is able to prevent lymphatic vessel formation.

The final antilymphatic hits, cinnarizine and flunarizine, are both diphenylpiperazine calcium channel antagonists. Flunarizine is a fluorine analogue of cinnarizine. Both drugs are classified as type IV, or nonselective, calcium channel antagonists (47). The fact that both flunarizine and cinnarizine were identified in our screen strongly suggests that this class of calcium channel antagonist has efficacy as an antilymphatic compound. A previous screen identified members of the class I calcium channel blockers, felodipine and nicardipine, as antilymphatic in Xenopus embryos, although these drugs did not inhibit lymphangiogenesis in in vitro assays (48). Neither felodipine nor nicardipine was present in our chemical library, but we did screen other class I calcium inhibitors (nimodipine and nifedipine). However, they did not display antilymphatic activity at our screening dose, suggesting that class IV calcium channel antagonists are more effective antilymphatic compounds than the class I inhibitors.

Flunarizine did not inhibit secondary sprouting in the zebrafish, but rather it caused the parachordal lymphangioblasts to undergo apoptosis. Like leflunomide, flunarizine does not inhibit human VEGFR activity at micromolar concentrations and therefore could be a useful agent to complement existing VEGFR3 inhibitors that, like kaempferol, target the early steps of lymphatic sprouting from veins. Although flunarizine was also able to inhibit mammalian lymphangiogenesis in a Matrigel plug assay, its efficacy as a potential antimetastatic agent was difficult to determine as it promoted tumor growth. Flunarizine has been shown to increase blood flow into tumors (49, 50). It is possible that increased tumor blood flow induced by flunarizine could account for the tumorigenic properties we observed. Our data suggest that flunarizine is not a suitable antilymphatic agent for the prevention of metastatic disease but it could still have utility in the treatment of other lymphatic-based pathologies such as the host rejection of corneal grafts.

This study has shown that zebrafish is a viable platform for the identification and development of mammalian antilymphatic compounds; however, we observed effects of the two drugs tested in our xenograft model that suggest they would not be useful in a tumor setting. This is not surprising given that we isolated antilymphatic compounds solely on the ability to prevent lymphangiogenesis, while the influence of these drugs on tumor growth and cancer cell migration was not assessed. Any future zebrafish antilymphatic screens should utilize additional secondary screens to ensure better translation of antilymphatic drugs in the prevention of metastasis. Possible secondary screens could ensure that antilymphatic compounds do not promote tumor growth or migration by conducting in vitro cancer cell line growth and migration assays.

No potential conflicts of interest were disclosed.

Conception and design: J.W. Astin, S.M.F. Jamieson, M.V. Flores, K.E. Crosier, P.S. Crosier

Development of methodology: J.W. Astin, S.M.F. Jamieson, M.V. Flores, P.S. Crosier

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): J.W. Astin, M.V. Flores, J.P. Misa, A. Chien

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): J.W. Astin, S.M.F. Jamieson, T.C.Y. Eng, K.E. Crosier

Writing, review, and/or revision of the manuscript: J.W. Astin, S.M.F. Jamieson, T.C.Y. Eng, K.E. Crosier, P.S. Crosier

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): T.C.Y. Eng, J.P. Misa, P.S. Crosier

Study supervision: S.M.F. Jamieson, M.V. Flores, K.E. Crosier, P.S. Crosier

This work was supported by grant number UOAX0813 from the New Zealand Ministry of Business, Innovation and Employment (to P.S. Crosier).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Nathanson
SD
. 
Insights into the mechanisms of lymph node metastasis
.
Cancer
2003
;
98
:
413
23
.
2.
Sundlisaeter
E
,
Dicko
A
,
Sakariassen
PO
,
Sondenaa
K
,
Enger
PO
,
Bjerkvig
R
. 
Lymphangiogenesis in colorectal cancer–prognostic and therapeutic aspects
.
Int J Cancer
2007
;
121
:
1401
9
.
3.
Alitalo
A
,
Detmar
M
. 
Interaction of tumor cells and lymphatic vessels in cancer progression
.
Oncogene
2012
;
31
:
4499
508
.
4.
Achen
MG
,
McColl
BK
,
Stacker
SA
. 
Focus on lymphangiogenesis in tumor metastasis
.
Cancer Cell
2005
;
7
:
121
7
.
5.
Christiansen
A
,
Detmar
M
. 
Lymphangiogenesis and cancer
.
Genes Cancer
2011
;
2
:
1146
58
.
6.
Dadras
SS
,
Paul
T
,
Bertoncini
J
,
Brown
LF
,
Muzikansky
A
,
Jackson
DG
, et al
Tumor lymphangiogenesis: a novel prognostic indicator for cutaneous melanoma metastasis and survival
.
Am J Pathol
2003
;
162
:
1951
60
.
7.
Kerjaschki
D
,
Regele
HM
,
Moosberger
I
,
Nagy-Bojarski
K
,
Watschinger
B
,
Soleiman
A
, et al
Lymphatic neoangiogenesis in human kidney transplants is associated with immunologically active lymphocytic infiltrates
.
J Am Soc Nephrol
2004
;
15
:
603
12
.
8.
Albuquerque
RJ
,
Hayashi
T
,
Cho
WG
,
Kleinman
ME
,
Dridi
S
,
Takeda
A
, et al
Alternatively spliced vascular endothelial growth factor receptor-2 is an essential endogenous inhibitor of lymphatic vessel growth
.
Nat Med
2009
;
15
:
1023
30
.
9.
He
Y
,
Rajantie
I
,
Pajusola
K
,
Jeltsch
M
,
Holopainen
T
,
Yla-Herttuala
S
, et al
Vascular endothelial cell growth factor receptor 3-mediated activation of lymphatic endothelium is crucial for tumor cell entry and spread via lymphatic vessels
.
Cancer Res
2005
;
65
:
4739
46
.
10.
Hirakawa
S
,
Brown
LF
,
Kodama
S
,
Paavonen
K
,
Alitalo
K
,
Detmar
M
. 
VEGF-C-induced lymphangiogenesis in sentinel lymph nodes promotes tumor metastasis to distant sites
.
Blood
2007
;
109
:
1010
7
.
11.
Hirakawa
S
,
Kodama
S
,
Kunstfeld
R
,
Kajiya
K
,
Brown
LF
,
Detmar
M
. 
VEGF-A induces tumor and sentinel lymph node lymphangiogenesis and promotes lymphatic metastasis
.
J Exp Med
2005
;
201
:
1089
99
.
12.
Skobe
M
,
Hawighorst
T
,
Jackson
DG
,
Prevo
R
,
Janes
L
,
Velasco
P
, et al
Induction of tumor lymphangiogenesis by VEGF-C promotes breast cancer metastasis
.
Nat Med
2001
;
7
:
192
8
.
13.
Caunt
M
,
Mak
J
,
Liang
WC
,
Stawicki
S
,
Pan
Q
,
Tong
RK
, et al
Blocking neuropilin-2 function inhibits tumor cell metastasis
.
Cancer Cell
2008
;
13
:
331
42
.
14.
Lin
J
,
Lalani
AS
,
Harding
TC
,
Gonzalez
M
,
Wu
WW
,
Luan
B
, et al
Inhibition of lymphogenous metastasis using adeno-associated virus-mediated gene transfer of a soluble VEGFR-3 decoy receptor
.
Cancer Res
2005
;
65
:
6901
9
.
15.
Roberts
N
,
Kloos
B
,
Cassella
M
,
Podgrabinska
S
,
Persaud
K
,
Wu
Y
, et al
Inhibition of VEGFR-3 activation with the antagonistic antibody more potently suppresses lymph node and distant metastases than inactivation of VEGFR-2
.
Cancer Res
2006
;
66
:
2650
7
.
16.
Alam
A
,
Blanc
I
,
Gueguen-Dorbes
G
,
Duclos
O
,
Bonnin
J
,
Barron
P
, et al
SAR131675, a potent and selective VEGFR-3-TK inhibitor with antilymphangiogenic, antitumoral, and antimetastatic activities
.
Mol Cancer Ther
2012
;
11
:
1637
49
.
17.
Hogan
BM
,
Herpers
R
,
Witte
M
,
Helotera
H
,
Alitalo
K
,
Duckers
HJ
, et al
Vegfc/Flt4 signalling is suppressed by Dll4 in developing zebrafish intersegmental arteries
.
Development
2009
;
136
:
4001
9
.
18.
Koltowska
K
,
Betterman
KL
,
Harvey
NL
,
Hogan
BM
. 
Getting out and about: the emergence and morphogenesis of the vertebrate lymphatic vasculature
.
Development
2013
;
140
:
1857
70
.
19.
Kuchler
AM
,
Gjini
E
,
Peterson-Maduro
J
,
Cancilla
B
,
Wolburg
H
,
Schulte-Merker
S
. 
Development of the zebrafish lymphatic system requires VEGFC signaling
.
Curr Biol
2006
;
16
:
1244
8
.
20.
Zon
LI
,
Peterson
RT
. 
In vivo drug discovery in the zebrafish
.
Nat Rev Drug Discov
2005
;
4
:
35
44
.
21.
Bussmann
J
,
Bos
FL
,
Urasaki
A
,
Kawakami
K
,
Duckers
HJ
,
Schulte-Merker
S
. 
Arteries provide essential guidance cues for lymphatic endothelial cells in the zebrafish trunk
.
Development
2010
;
137
:
2653
7
.
22.
Tran
TC
,
Sneed
B
,
Haider
J
,
Blavo
D
,
White
A
,
Aiyejorun
T
, et al
Automated, quantitative screening assay for antiangiogenic compounds using transgenic zebrafish
.
Cancer Res
2007
;
67
:
11386
92
.
23.
Wang
C
,
Tao
W
,
Wang
Y
,
Bikow
J
,
Lu
B
,
Keating
A
, et al
Rosuvastatin, identified from a zebrafish chemical genetic screen for antiangiogenic compounds, suppresses the growth of prostate cancer
.
Eur Urol
2010
;
58
:
418
26
.
24.
Buchanan
CM
,
Shih
JH
,
Astin
JW
,
Rewcastle
GW
,
Flanagan
JU
,
Crosier
PS
, et al
DMXAA (Vadimezan, ASA404) is a multi-kinase inhibitor targeting VEGFR2 in particular
.
Clin Sci
2012
;
122
:
449
57
.
25.
Flores
MV
,
Hall
CJ
,
Crosier
KE
,
Crosier
PS
. 
Visualization of embryonic lymphangiogenesis advances the use of the zebrafish model for research in cancer and lymphatic pathologies
.
Dev Dyn
2010
;
239
:
2128
35
.
26.
Okuda
KS
,
Astin
JW
,
Misa
JP
,
Flores
MV
,
Crosier
KE
,
Crosier
PS
. 
lyve1 expression reveals novel lymphatic vessels and new mechanisms for lymphatic vessel development in zebrafish
.
Development
2012
;
139
:
2381
91
.
27.
Lawson
ND
,
Weinstein
BM
. 
In vivo imaging of embryonic vascular development using transgenic zebrafish
.
Dev Biol
2002
;
248
:
307
18
.
28.
Lam
EY
,
Hall
CJ
,
Crosier
PS
,
Crosier
KE
,
Flores
MV
. 
Live imaging of Runx1 expression in the dorsal aorta tracks the emergence of blood progenitors from endothelial cells
.
Blood
2010
;
116
:
909
14
.
29.
Kaufman
CK
,
White
RM
,
Zon
L
. 
Chemical genetic screening in the zebrafish embryo
.
Nat Protoc
2009
;
4
:
1422
32
.
30.
Astin
JW
,
Haggerty
MJ
,
Okuda
KS
,
Le Guen
L
,
Misa
JP
,
Tromp
A
, et al
Vegfd can compensate for loss of Vegfc in zebrafish facial lymphatic sprouting
.
Development
2014
;
141
:
2680
90
.
31.
Huber
S
,
Bruns
CJ
,
Schmid
G
,
Hermann
PC
,
Conrad
C
,
Niess
H
, et al
Inhibition of the mammalian target of rapamycin impedes lymphangiogenesis
.
Kidney Int
2007
;
71
:
771
7
.
32.
Kobayashi
S
,
Kishimoto
T
,
Kamata
S
,
Otsuka
M
,
Miyazaki
M
,
Ishikura
H
. 
Rapamycin, a specific inhibitor of the mammalian target of rapamycin, suppresses lymphangiogenesis and lymphatic metastasis
.
Cancer Sci
2007
;
98
:
726
33
.
33.
Schulz
MM
,
Reisen
F
,
Zgraggen
S
,
Fischer
S
,
Yuen
D
,
Kang
GJ
, et al
Phenotype-based high-content chemical library screening identifies statins as inhibitors of in vivo lymphangiogenesis
.
Proc Natl Acad Sci U S A
2012
;
109
:
E2665
74
.
34.
Bartlett
RR
,
Dimitrijevic
M
,
Mattar
T
,
Zielinski
T
,
Germann
T
,
Rude
E
, et al
Leflunomide (HWA 486), a novel immunomodulating compound for the treatment of autoimmune disorders and reactions leading to transplantation rejection
.
Agents Actions
1991
;
32
:
10
21
.
35.
Davis
JP
,
Cain
GA
,
Pitts
WJ
,
Magolda
RL
,
Copeland
RA
. 
The immunosuppressive metabolite of leflunomide is a potent inhibitor of human dihydroorotate dehydrogenase
.
Biochemistry
1996
;
35
:
1270
3
.
36.
Schledzewski
K
,
Falkowski
M
,
Moldenhauer
G
,
Metharom
P
,
Kzhyshkowska
J
,
Ganss
R
, et al
Lymphatic endothelium-specific hyaluronan receptor LYVE-1 is expressed by stabilin-1+, F4/80+, CD11b+ macrophages in malignant tumors and wound healing tissue in vivo and in bone marrow cultures in vitro: implications for the assessment of lymphangiogenesis
.
J Pathol
2006
;
209
:
67
77
.
37.
Calderon-Montano
JM
,
Burgos-Moron
E
,
Perez-Guerrero
C
,
Lopez-Lazaro
M
. 
A review on the dietary flavonoid kaempferol
.
Mini Rev Med Chem
2011
;
11
:
298
344
.
38.
Cui
Y
,
Morgenstern
H
,
Greenland
S
,
Tashkin
DP
,
Mao
JT
,
Cai
L
, et al
Dietary flavonoid intake and lung cancer–a population-based case-control study
.
Cancer
2008
;
112
:
2241
8
.
39.
Nothlings
U
,
Murphy
SP
,
Wilkens
LR
,
Henderson
BE
,
Kolonel
LN
. 
Flavonols and pancreatic cancer risk: the multiethnic cohort study
.
Am J Epidemiol
2007
;
166
:
924
31
.
40.
Gates
MA
,
Tworoger
SS
,
Hecht
JL
,
De Vivo
I
,
Rosner
B
,
Hankinson
SE
. 
A prospective study of dietary flavonoid intake and incidence of epithelial ovarian cancer
.
Int J Cancer
2007
;
121
:
2225
32
.
41.
Luo
H
,
Rankin
GO
,
Juliano
N
,
Jiang
BH
,
Chen
YC
. 
Kaempferol inhibits VEGF expression and in vitro angiogenesis through a novel ERK-NFkappaB-cMyc-p21 pathway
.
Food Chem
2012
;
130
:
321
8
.
42.
Luo
H
,
Rankin
GO
,
Liu
L
,
Daddysman
MK
,
Jiang
BH
,
Chen
YC
. 
Kaempferol inhibits angiogenesis and VEGF expression through both HIF dependent and independent pathways in human ovarian cancer cells
.
Nutr Cancer
2009
;
61
:
554
63
.
43.
McLean
JE
,
Neidhardt
EA
,
Grossman
TH
,
Hedstrom
L
. 
Multiple inhibitor analysis of the brequinar and leflunomide binding sites on human dihydroorotate dehydrogenase
.
Biochemistry
2001
;
40
:
2194
200
.
44.
Williamson
RA
,
Yea
CM
,
Robson
PA
,
Curnock
AP
,
Gadher
S
,
Hambleton
AB
, et al
Dihydroorotate dehydrogenase is a high affinity binding protein for A77 1726 and mediator of a range of biological effects of the immunomodulatory compound
.
J Biol Chem
1995
;
270
:
22467
72
.
45.
Kaplan
MJ
. 
Leflunomide Aventis Pharma
.
Curr Opin Investig Drugs
2001
;
2
:
222
30
.
46.
White
RM
,
Cech
J
,
Ratanasirintrawoot
S
,
Lin
CY
,
Rahl
PB
,
Burke
CJ
, et al
DHODH modulates transcriptional elongation in the neural crest and melanoma
.
Nature
2011
;
471
:
518
22
.
47.
Opie
LH
,
Buhler
FR
,
Fleckenstein
A
,
Hansson
L
,
Harrison
DC
,
Poole-Wilson
PA
, et al
International society and federation of cardiology: working group on classification of calcium antagonists for cardiovascular disease
.
Am J Cardiol
1987
;
60
:
630
2
.
48.
Kalin
RE
,
Banziger-Tobler
NE
,
Detmar
M
,
Brandli
AW
. 
An in vivo chemical library screen in Xenopus tadpoles reveals novel pathways involved in angiogenesis and lymphangiogenesis
.
Blood
2009
;
114
:
1110
22
.
49.
Kaelin
WG
 Jr
,
Shrivastav
S
,
Jirtle
RL
. 
Blood flow to primary tumors and lymph node metastases in SMT-2A tumor-bearing rats following intravenous flunarizine
.
Cancer Res
1984
;
44
:
896
9
.
50.
Dewhirst
MW
,
Ong
ET
,
Madwed
D
,
Klitzman
B
,
Secomb
T
,
Brizel
D
, et al
Effects of the calcium channel blocker flunarizine on the hemodynamics and oxygenation of tumor microvasculature
.
Radiat Res
1992
;
132
:
61
8
.

Supplementary data