Abstract
Fumagillin, an unstable anti-angiogenesis mycotoxin, was synthesized into a stable lipase-labile prodrug and incorporated into integrin-targeted lipid-encapsulated nanoparticles (αvβ3-Fum-PD NP). Dual anti-angiogenic therapy combining αvβ3-Fum-PD NP with zoledronic acid (ZA), a long-acting osteoclast inhibitor with proposed anti-angiogenic effects, was evaluated. In vitro, αvβ3-Fum-PD NP reduced (p<0.05) endothelial cell viability and tube formation without impacting macrophage viability. ZA suppressed (p<0.05) macrophage viability at high dosages in vitro but not endothelial cell proliferation. 3D MRI neovascular imaging of 17d rabbit Vx2 tumors showed no effect with ZA, whereas αvβ3-Fum-PD NP alone and with ZA decreased angiogenesis (p<0.05). Immunohistochemistry revealed decreased (p<0.05) microvascularity with αvβ3-Fum-PD NP and ZA and further microvascular reduction (p<0.05) with the dual-therapy. However, in vivo, ZA did not decrease tumor macrophage numbers nor cancer cell proliferation, whereas αvβ3-Fum-PD-NPs reduced both measures. Dual-therapy with ZA and αvβ3-Fum-PD-NP may provide enhanced neo-adjuvant utility if macrophage ZA uptake is increased.
Keywords: fumagillin, zoledronic acid, cancer, nanoparticle, MRI, molecular imaging
Graphical Abstract

Schematic representation of the activated integrin αvβ3-targeted perfluorocarbon nanoparticle fumagillin prodrug (αvβ3-Fum-PD) chemistry, contact facilitated drug delivery (CFDD) mechanism, and lipase enzymatic liberation of active fumagillol.
Background
Angiogenesis is required for tumor growth beyond a certain size and is an attractive therapeutic target. Current anti-angiogenic agents directly target the tumor endothelium through inhibition of the VEGF pathway. Tumors circumvent anti-VEGF blockade in part through alternative pro-angiogenic factors and endothelial signaling pathways 1. Angiogenesis is a complex process that involves multiple host cell types including endothelial cells, myeloid derived macrophages, myeloid derived suppresser cells (MDSCs) and T-cells 2, 3. Tumor associated macrophages (TAMs) promote angiogenesis through soluble factors that increase endothelial cell proliferation and matrix degradation 4. Macrophage driven angiogenesis significantly contributes to resistance to VEGF pathway inhibitors 1, 4. Thus, successful anti-angiogenesis treatment strategies for unresponsive and resistant tumor types will likely require combination therapies targeting multiple pathways driving neovasculature expansion.
Fumagillin, a hydrophobic mycotoxin produced by Aspergillus fumigatus, has been demonstrated to suppress angiogenesis by inhibition of methionine aminopeptidase 2 (MetAP2) 5–7. Of two methionine aminopeptidase forms in eukaryotes, only MetAP2 is up-regulated during cellular proliferation, providing greater efficiency (1000-fold) catalyzing methionine removal from certain proteins, such as glyceraldehydes-3-phosphate, which can modulate subsequent activity, stability or subcellular localization8–10. TNP-470 is a water-soluble, stable form of fumagillin that was demonstrated in a variety of rodent cancer models, 11–13 but when tested in human clinical trials, 14–16 the molecule possessed significant liabilities including a brief systemic half-life and moderately severe symptoms of neurotoxicity14.
We have reported effective in vivo neovascular delivery of native fumagillin with αvβ3-targeted perfluorocarbon (PFC) nanoparticles at a fraction of the dosage that was required systemically for TNP-470 in clinical studies 17–19. Fumagillin is very hydrophobic and dissolves easily into the phospholipid membrane component of PFC nanoparticles. When targeted to angiogenic endothelial cells, it was efficiently delivered through a mechanism we described as “contact facilitated drug delivery” (CFDD) 20–22. Although αvβ3-targeted perfluorocarbon (PFC) nanoparticle delivery of native fumagillin was effective in preclinical animal models, simultaneous pharmacokinetic tracking of individual nanoparticle components revealed that much of the compound was prematurely lost in blood before the particles bound to the neovasculature 23. To improve the light and chemical stability of fumagillin and to increase its retention in lipid-encapsulated nanoparticles during circulation to target tissues, an Sn-2 lipase labile phospholipid fumagillin prodrug (Fum-PD) was designed, synthesized and incorporated stably into the surfactant of αvβ3-targeted perfluorocarbon (PFC) nanoemulsions (Fig 1) 23. Early data in vivo suggest that αvβ3-Fum-PD PFC NP were effective in mouse Matrigel™ angiogenesis plug and in an inflammatory arthritis model, but its effectiveness in cancer, particularly the aggressive rabbit Vx2 tumor model, is unknown 24, 25.
Figure 1.
Schematic representation of the activated integrin αvβ3-targeted perfluorocarbon nanoparticle fumagillin prodrug (αvβ3-Fum-PD) chemistry, contact facilitated drug delivery (CFDD) mechanism, and lipase enzymatic liberation of active fumagillol.
Zoledronic acid (ZA) is a long-acting amino-bisphosphonate approved to treat osteoporosis and to prevent cancer-related bone fracture and pain. Recent clinical evidence suggests that ZA may have beneficial effects on primary breast cancer growth and patient survival 26–29. Cancer bearing mice treated with ZA have reduced vasculature through an unresolved mechanism that implicates a complex interplay among MDSC, TAMs, tumor cells and T-cells 30, 31. In patients, ZA treatment correlated with reduced circulating angiogenic factors including VEGF and PDGF, but a clear effect on the tumor vasculature was not demonstrated. Importantly, myeloid lineage osteoclasts and macrophages have the ability for ZA endocytic uptake and can phagocytose ZA bound to bone or tumor microcalcifications 30, 32, 33. We hypothesized that the long-acting nature of ZA via myeloid mediated pathway would complement the anti-angiogenesis benefits of αvβ3-Fum-PD PFC NP nanoparticles in cancer.
The objectives of these studies were: 1) to characterize the anti-angiogenesis effectiveness of αvβ3-Fum-PD in vitro and in an aggressive large animal cancer model; 2) to determine and characterize the anti-neovascular effects of intravenous ZA in the Vx2 rabbit model; and 3) to determine if dual therapy with intravenous ZA and αvβ3-Fum-PD nanoparticles offer complementary therapeutic benefit in cancer.
Methods
Primary cell assays
Primary human umbilical vein endothelial cells (HUVECS) obtained from Lonza (07/01/13) expressed CD31/105, von Williebrand Factor VIII, and were positive for acetyated low density lipoprotein uptake. HUVEC were grown to 70% confluence in VascuLife Endothelial Cell Culture Media (Lifeline Cell Technologies, Fredrick, MD, USA) and maintained for <5 passages. Bone marrow macrophage (BMM) cultures were isolated from C57Bl/6J mice, plated and maintained in macrophage media culture for 3 days (MEM alpha, Life Technologies + 10% FBS + and 100ng/mL M-CSF) 34. Osteoclasts were cultured 6 days in macrophage media (described above) + RANKL (50ng/ml) following isolation of BMMs 35, 36. Cell viability was measured using MTT assay (Sigma-Aldrich, St. Louis, MO, USA)35. To assess endothelial tube formation, HUVEC (100μl; 30,000 cells/well) were plated in tissue culture plates (96-well) coated with 100μl Matrigel™ (BD Biosciences, San Jose, CA, USA) and incubated at 37°C for 16hr. For nanoparticle binding assays, HUVEC were plated for 24 hours, washed with PBS, then incubated with fluorescent αvβ3-targeted or non-targeted NPs (1hr, 37 C) with gentle continuous shaking. After removal of unbound particles with multiple PBS washes, cells were recovered with trypsin and analyzed by flow cytometry (BD LSRFortessa, BD Biosciences, San Jose, CA, USA).
To assess Vx2 tumor composition, minced tumors were incubated in 1X collagenase/hyaluronidase solution (STEMCELL Technologies, Vancouver, BC, Canada) for 4hr shaking at room temperature. Single cell suspensions were isolated by filtration (BD Falcon 70μM cell strainer, Fisher Scientific, Pittsburg, PA, USA) and incubated with fluorescent anti-mouse pan-cytokeratin (AE1/A13, 1:200, ebioscience, San Diego, CA, USA) and anti-mouse CD11b (M1/70, 1:400, eBioscience, San Diego, CA, USA) antibodies for 30 minutes. Unbound antibodyy by excess PBS washes then cells were characterized by flow cytometry (BD LSRFortessa, BD Biosciences, San Jose, CA, USA).
Quantitative reverse-transcription PCR
RNA was extracted using RNeasy Mini kit (Qiagen, Venlo, Netherlands) and cDNA generated using iScript (Bio-Rad, Hercules, CA, USA). Quantitative PCR was completed using SsoFast EVA Green Supermix (Bio-Rad). Primer sequences were CD68 forward- CTCCACCTCGACCTGCTCT and CD68 reverse- ATGATGAGGGGCACCAAGAT.
Transwell co-culture assays
BMMs (20,000/well) were cultured as described on 24-well polycarbonate transwell inserts (0.4um pores, Fisher Scientific, Pittsburg, PA, USA). After 24hr media was aspirated and replaced with fresh media containing mouse IL-4 (5ng/ml) to polarize to an M2 phenotype. Twenty-four hours later this media was replaced with macrophage media + IL-4 (5ng/ml) + LPS (100ng/ml) for 6hr. Transwells were washed with PBS and transferred to 24-well plates containing HUVECs (30,000/well). Viability was measured by MTT assay (Sigma-Aldrich).
Vx-2 rabbit tumor model
Animal studies were performed under a protocol approved and monitored by the Washington University Animal Studies Committee. Male New Zealand White rabbits (~2 kg) were anesthetized with intramuscular ketamine and xylazine. The Vx-2 tumor (Division of Cancer Treatment and Diagnosis Tumor Repository, National Cancer Institute, USA) was implanted as described previously 37. All animal care and experimental protocols were approved and conducted in accordance with Washington University guidelines.
Synthesis of Sn2 lipase labile fumagillin-prodrug
An Sn2 fumagillin-prodrug (Fum-PD) was envisioned and the compound was synthesized in a straightforward way in two-steps involving saponification of fumagillin dicyclohexylamine salt to fumagillol and a subsequent esterification with oxidized lipid 1-palmitoyl-2-azelaoyl-sn-glycero-3-phosphocholine 23. (See Supplemental Materials for detail
αvβ3-Targeted perfluorocarbon nanoparticle synthesis incorporating fumagillin-prodrug
An αvβ3-integrin antagonist coupled to phosphatidylethanolamine-PEG2000 was used for neovasculature homing (gift from Kereos, Inc, St. Louis, MO, USA). This quinolone nonpeptide antagonist developed by Bristol-Myers Squibb Medical Imaging to αvβ3-integrin (US patent 6,511,648 and related patents) was initially reported and characterized as the 111In-DOTA conjugate RP748 and cyan 5.5 homologue TA145. The targeted particles presented ~300 ligands/particle with an IC50 of 50 pM for the Mn2+-activated αvβ3-integrin 19. Homing specificity to neovascular sprouts was previously demonstrated in a well defined in Matrigel™ plug study 38.
αvβ3-Fum-PD perfluorocarbon (PFC) nanoparticles (NP) were prepared as a microfluidized suspension of 20% (v/v) perfluoroctylbromide (Exfluor Inc., Round Rock, TX, USA), 2.0% (w/v) of a surfactant co-mixture, and 1.7% (w/v) glycerin. The surfactant co-mixture of NP included: 97.6 mole% lecithin, 0.15 mole% of αvβ3-ligand conjugated lipid and 2.3 mole% of the Fum-PD. The surfactant components were combined with the PFC, deionized water, and glycerin with pH adjusted to 6.5 with 5% carbonate buffer. The mixture was pre-blended (Tissumizer Mark II, Tekmar, Cincinnatti, Ohio, USA) then homogenized at 20,000 psi for 4 minutes (M110s, Microfluidics Inc., Westwood, MA, USA). Routine NP characterization revealed: nominal size of 227.2 nm, polydispersity of 0.072, and zeta potential of −10.74 mV (Brookhaven Instruments Co, Holtsville, NY, USA) (Fig 1).
Synthesis of αvβ3-targeted manganese oleate nanocolloids with gadolinium chelate
For MR imaging, paramagnetic manganese oleate (MnOL) nanocolloid (NC) with a very low concentration of lipid-anchored-chelated gadolinium was synthesized to produce αvβ3-MnOL-Gd NC. 39, 40 Briefly, Mn-oleate was suspended in almond oil to form the emulsion core, which was encapsulated with phospholipid surfactant via microfluidization to produce the base nanocolloid. The surfactant mixture was comprised of 0.15 mole% of αvβ3-ligand conjugated lipid, 1.25 mole% Gd-DOTA-PE (gadolinium-(1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid)- phosphatidylethanolamine) and phosphatidylcholine (PC). In selected preparations, the lipid surfactant included Texas Red dye coupled to caproyl-phosphatidylethanolamine (Life Technologies; Avanti Polar Lipids, Alabaster, Alabama, USA) at 0.5 mol % for microscopic fluorescence imaging. Nanocolloids were extensively dialyzed against water using a 20,000 Da MWCO cellulose membrane, passed through a 0.45 μm syringe filter, and stored under argon atmosphere at 4°C to reduce oxidation and bacterial growth. Nanocolloid characterization revealed a nominal particle size of 162.5nm, polydispersity of 0.071, zeta potential of −21.62mV. NC were preserved under inert gas in sterile sealed vials until use. Full physical-chemical, MR characterization, pharmacokinetics, biodistribution and bioelimination, and demonstration of neovascular imaging with αvβ3-MnOL-Gd NC was previously reported. 39, 40
MR neovascular molecular imaging
On day 17 post tumor implantation, rabbits were anesthetized with 1% to 2% Isoflurane in oxygen to effect and imaged in a clinical 3T MR scanner (Philips Achieva, Andover, MA, USA) with an 8-element SENSE coil using a high-resolution, T1-weighted, fat suppressed, 3D gradient echo sequence (TR/TE = 42.6/6.1ms, 45° flip angle, 2 NSA, 0.25 × 0.25 × 0.5 mm resolution, SPIR fat suppression). Images were obtained before and 2.5 hours after IV administration of αvβ3-MnOL-Gd NC (1ml/kg) to compare the influence of the treatments on angiogenesis and tumor volumes.
Signal intensities of MR images were analyzed with custom MATLAB software (The Math Works, Natick, MA, USA). Dynamic T1-weighted signal intensities were normalized to a Gd-DTPA doped water reference standard included within each imaging field-of-view. For each animal, a region-of-interest (ROI) was manually placed around the tumor edge on each baseline slice, and the standard deviation of the average tumor signal at baseline was calculated. Voxels on post-injection MRI images were defined as enhanced if the signal intensity exceeded the mean tumor signal at baseline by more than two standard deviations. The tumor ROI was automatically partitioned into a rim and core region using an automated erosion method (MATLAB). The core region was set to half of the ROI volume. The volume of enhancement was calculated, as well as the relative distribution of enhancing voxels in the periphery versus the core. High-resolution three dimensional (3D) tumor reconstructions were created in MATLAB to map the spatial distribution of the enhancing neovasculature in each treatment group. The overall 3D structure was displayed as a mesh surface plot using iso-surface rendering and a smoothing filter. A surface plot of the enhancing voxels was reconstructed similarly, and overlaid onto the tumor volume.
MicroCT
To corroborate the known effect of ZA on bone, rabbit femurs were suspended in agarose and femoral epiphyses were scanned by micro–computed tomography (mCT-40; Scanco Medical, Wayne, PA, USA). For image acquisition, femurs were scanned in a 30mm sample holder. Fifty1.5mm image slices of trabecular bone beginning 1.2mm from the epiphyseal growth plate were acquired. A 3D cubical voxel model of each bone was built using contours drawn within the cortical shell, and calculations were made for trabecular thickness (Tb.Th) as described previously 41, 42.
Histology
Rabbit tumors were harvested at study endpoint and embedded in OCT compound. Frozen sections were stained for immunohistochemistry (IHC) with mouse anti-human CD31+ (DAKO-M0823, 1:2000, DAKO North America, Carpinteria, CA, USA) and proliferating cellular nuclear antigen (PCNA, ab29, 1:2000, Abcam, Cambridge, MA, USA). Whole tumor slices (n=4) were quantified due to significant morphological heterogeneity. PCNA, proliferating cell nuclear antigen, was quantified as number of positive cells/whole tumor area/slice. CD31 was quantified along the entire tumor rim (defined as non-necrotic counterstained regions adjacent to the surrounding stroma) and reported as area of positive stain/ROI area. Samples were quantified with Visiopharm software (Visiopharm, Broomfield, CO, USA) and threshold levels were set equally across all samples to exclude background staining. Vx2 tumors were immunostained with anti-mouse CD11b (M1/70, 1:200, eBioscience, San Diego, CA, USA) and quantified along the tumor rim. Three ROI’s per sample were randomly selected and the number of CD11b+ cells was counted manually.
Statistical analysis
In vitro experiments were analyzed by t-test or one-way ANOVA with a Bonferroni post-hoc test using PRISM software, (GraphPad, La Jolla, CA, USA) as appropriate. All in vitro experiments were repeated 2–3 times and representative experiments are shown as mean ± SEM. In vivo imaging results were analyzed with SAS (Cary, NC, USA) using ANOVA and Tukey’s range test. Significant differences between mean were declared at p<0.05 or less.
Results
In vitro effects of zoledronic acid and fumagillin on endothelial and macrophage-osteoclast viability
Fumagillin Sn2 prodrug (Fum-PD) and native fumagillin (Fum) were compared in HUVEC culture as equimolar titrations of the active compound. Both Fum and Fum-PD in DMSO produced nearly identical dose-related decreases in HUVEC proliferation in culture. Although Fum-PD required endothelial cell membrane solubility and subsequent cytosolic enzymatic liberation of active drug (API) from its phospholipid backbone, its net bioactivity was unchanged from native free drug. Further, the monotonic decrease in cell proliferation with increasing dosage of Fum-PD indicated that neither the rate of cellular uptake nor cytosolic API liberation limited the prodrug’s effectiveness. (Figure 2A) Similarly, Fum-PD in DMSO inhibited endothelial tube formation in 3D culture further corroborating the anti-angiogenic biopotency of the photochemically stabilized compound (Supplementary Figure 1).
Figure 2.
Integrin αvβ3-targeted fumagillin prodrug (Fum-PD) nanoparticles (αvβ3-Fum-PD NP) reduced endothelial cell viability. A) Dose-dependent viability of HUVEC exposed to native fumagillin (Fum) or Fum-PD dissolved in DMSO and assessed using the MTT assay after 48hr drug exposure. B) Flow cytometry of HUVEC incubated with fluorescent (Texas Red) αvβ3-NP (black) compared to fluorescent non-targeted NP (grey) and no NP controls (white). C) Viability of HUVEC by MTT assay 48hr after 1hr treatment (**p<0.01, ***p<0.001). D) Viability of primary macrophages by MTT assay after 48hr exposure with Fum-PD in DMSO.
Fluorescent nontargeted and αvβ3-Fum-PD NP (i.e., 597μM, 0.275mg/ml fumagillin API) were incubated with activated HUVECs. Flow cytometry demonstrated a marked increase in integrin-cell binding whereas the nontargeted control had little cellular affinity. (Figure 2B) Subsequently, αvβ3-Fum-PD NP and control NP were incubated with HUVEC at final doses of 100, 200, and 300 pM, which reduced (p<0.01) endothelial viability in a dose-dependent manner for the targeted therapeutic but not for the nontargeted nanoparticles. (Figure 2C) These results demonstrated that αvβ3-Fum-PD NP drug delivery was efficacious relative to the nontargeted control particles, which remained 100% viable. Unlike the administration of Fum-PD in DMSO, the endothelial cell uptake of αvβ3-Fum-PD NP was dependent on the number of particles binding to the cells, which reflected the density of cell surface activated αvβ3-integrin available. In contradistinction to endothelial cells, the viability of activated primary macrophages was unaffected by Fum-PD in DMSO (Figure 2D). Although activated macrophages express αvβ3-integrin and may be targeted by αvβ3-Fum-PD NP, the mycotoxin did not effect macrophage viability at concentrations that decreased HUVEC viability by greater than 80%.
Titrated levels of ZA had little effect on HUVEC viability except at the highest dosage level (50μM). (Figure 3A) Moreover, ZA added to HUVEC cultures did not further decrease the viability achieved with Fum-PD. (Figure 3B) Analogous results were appreciated for endothelial tube formation. HUVEC proliferation in culture was enhanced by co-culture with M2 polarized macrophages (tumor promoting). (Figure 3C) ZA reduced macrophage viability slightly at concentrations up to 10 μM (p>0.05), but beyond this dose threshold, macrophage viability decreased precipitously (p<0.05). Similar findings were noted at higher concentrations in primary osteoclasts in culture, corroborating this result. (Figure 3D, Supplemental Figure 2) These data illustrate the supportive influence of macrophages on proliferating endothelial cell viability. Overall, ZA had minimal effect of HUVEC in culture compared with myeloid cells treated equivalently and the significant effectiveness against macrophage viability observed depended upon the direct exposure at concentrations exceeding 10μM.
Figure 3.
Zoledronic acid (ZA) reduced viability of myeloid lineage cells. A) HUVEC viability by MTT after 48hr exposure to titrated ZA concentrations. B) MTT assay for HUVEC after 48hr exposure to saline, ZA (25μM), Fum-PD (1μM) and ZA+ Fum-PD (25μm and 1μm respectively). C) MTT assay for viability of HUVECs alone or co-cultured with M2 polarized macrophages for 24hr D) Viability of macrophages after 48hr ZA exposure. *p<0.05 and ***p<0.001.
αvβ3 Fum-PD NPs and zoledronic acid decrease tumor vasculature and macrophages in Vx2 rabbit tumors
The potential combination of αvβ3-Fum-PD NP and ZA was evaluated in an aggressive syngeneic immunocompetent rabbit Vx2 squamous cell carcinoma tumor model. This tumor has significant progressive neovascular expansion that was serially documented with MR neovascular molecular imaging using αvβ3-paramagnetic PFC nanoparticles at 3T 43. In the present study, rabbits were distributed into 4 treatment groups (n=8/treatment) and received either: intravenous saline, ZA, αvβ3-Fum-PD NPs, and ZA plus αvβ3 Fum-PD NPs. Animals were dosed with αvβ3-Fum-PD NP (~0.28 mg Fum/kg) on days 9, 12 and 15 post-implantation. ZA (40μg/kg) was given intravenously on days 5 and 12. The fumagillin dose was over 100-fold lower than the TNP-470 treatments used in human clinical studies 15, 44, 45. The ZA dosage (40 mg/kg IV) was equivalent to current clinical regimens used for breast cancer patients with metastatic bone disease. MicroCT of rabbit femurs confirmed that the ZA dosing regimen increased trabecular thickness compared to saline controls, as it is clinically intended to do. (Figure 4A,B)
Figure 4.
Zoledronic acid (ZA) increased trabecular thickness. A) Femur trabecular bone thickness (n=5). B) Representative images of microCT reconstruction of rabbit femur exposed to saline versus ZA IV, scale bar = 5mm.
On day 17, αvβ3-MnOL-Gd nanocolloids were used for MR neovascular molecular imaging in cancer for the first time. As previously appreciated with the Vx2 tumor model, 3D MR neovascular map revealed an extensive heterogeneous and asymmetrical distribution of neovascular expansion predominantly around the tumor periphery. (Figure 5A,B) αvβ3-Fum-PD NP markedly decreased (p<0.05) Vx2 tumor neovascularity relative to the control, whereas, ZA therapy elicited only a trend (p>0.05) toward decreased neovascular expansion. (Figure 5A,B) In combination with αvβ3-Fum-PD NP, ZA did not further decrease (p>0.05) the MR molecular imaging signal achieved. While differences in neovascular density are quantifiable with MR molecular imaging at a single point in time, they may not reflect cumulative vascular changes over the treatment course.
Figure 5.
3D MR neovascular mapping with αvβ3-MnOL-Gd NC in 17d Vx2 tumors following saline, ZA, αvβ3-Fum-PD NP, or αvβ3-Fum-PD NP plus ZA. A) Surface volume percentage of neovascular voxels in the outer tumor (rim) for each treatment group (n=8, *p<0.05). B) Representative images of 3D MRI neovascular reconstructed maps with purple representing neovascular rich voxels overlaid onto chain-mesh surface-rendered Vx2 tumor volume.
Microscopic immunohistochemistry (CD31+) was used to evaluate overall vascular changes in the tumor periphery using CD31+, an endothelial cell marker expressed by forming and established blood vessels. Microscopically, both ZA and αvβ3-Fum-PD NP therapy significantly decreased (p<0.05) tumor peripheral microvascularity versus controls. (Figure 6A,B) Further, the combination of ZA and αvβ3-Fum-PD NP additively decreased tumor microvascularity further (p<0.05). (Figure 6A)
Figure 6.
Effect of zoledronic acid (ZA) and αvβ3-targeted fumagillin prodrug nanoparticle (αvβ3-Fum-PD NP) therapy on microvasculature (CD31+) in the Vx2 rabbit tumor model. A) Percent CD31+ stain quantified from tumor immunohistochemistry (n=4, *p<0.05). The area of CD31+ stain was normalized to the total tumor area in the region of interest B) Representative images of CD31+ staining, scale bar = 0.5mm. C) Number of PCNA+ cells in Vx2 tumor tissue by immunohistochemistry (n=4). The number of PCNA expressing cells was normalized to the total area of each tumor. D) Representative images of PCNA stained tumors, scale bar = 11mm (*P<0.05).
Decreased tumor vascularity, whether achieved with ZA and αvβ3-Fum-PD NP, was not associated with decreased tumor volume by estimated by MR and corroborated closely by the dissected tumor weight. (Supplementary Figure 3). Vx2 tumor cell proliferation, as reflected by PCNA staining, was reduced by αvβ3-Fum-PD NP (p<0.05) (Figure 6C,D). In contradistinction, ZA did not impact cancer cell proliferation despite its measurable diminishment of tumor vascularity. Moreover, the reduced tumor cell proliferation achieved with αvβ3-Fum-PD NP was not increased by ZA despite the additive anti-vascular effects noted microscopically. αvβ3-Fum-PD NP diminished tumor viability more effectively through acute transient depressions of tumor neovascularity or by collateral effects of endothelial apoptosis than the slower chronic reductions in vascularity elicited by ZA through inhibition of macrophage activity.
Control Vx2 tumor tissue was primarily composed of pan-cytokeratin+ tumor cells (75%) and CD11b+ macrophage infiltrate. (19%, Figure 7A) Similar to the prevalence of microcalcifications in a small cohort of human breast tumors examined (7/7), all Vx2 tumor (16/16) stained positively for calcium microdeposits, which could have provided substantial substrate for ZA binding and subsequent TAM phagocytosis. (Figure 7B,C) The density of rabbit tumor microcalcification grossly exceeded the levels observed in the human breast cancer sample and likely did not limit this route of delivery. Nevertheless, IV ZA elicited no significant reduction in macrophage CD68 expression (Figure 7D), measured by qPCR, while αvβ3-Fum-PD NP decreased (p<0.05) in CD68 expression in the tumors.
Figure 7.
Characterization of Vx2 major pathological features. A) Analysis of relative tumor cell (pan –cytokeratin) and macrophage (CD11b) cell population distribution in the Vx2 tumors using flow cytometry. B) Von Kossa stained (black) microcalcifications in control Vx2 tumor, scale bar = 250μm (top) and 100μm (bottom) and C) in excised human breast cancers, scale bar = 100μm. D) mRNA expression of macrophage marker CD68 in Vx2 tumors by qPCR (*p<0.05). E) Number of CD11b+ cells per field of view (FOV) at the tumor rim in the Vx2 tumor tissue by immunofluorescence (n=4, *p<0.05). F) Representative images of CD11b+ staining, scale bar = 100μm.
Interestingly, ZA reduced CD11b+ leukocyte number at the tumor rim (p<0.05), while αvβ3-Fum-PD NP elicited no reduction in CD11b+ cells and possibly an increase. Of note, anti-angiogenesis treatments have been suggested to overcome tumor endothelial anergy, angiogenic factor induced suppression of adhesion molecules on neovascular endothelial cells, which has been observed as a marked increase in tumor associated CD8+ lymphocytes in the Vx2 model 46–49, the leukocyte – tumor story remains complex and will require continued focused research.
Discussion
In this study, fumagillin Sn2 lipase-labile prodrug was incorporated into the surfactant of αvβ3-perfluorocarbon nanoparticles and shown to be an effective anti-angiogenic therapy in the Vx2 rabbit tumor model. αvβ3-Fum-PD-NP decreased tumor angiogenesis as demonstrated by MR molecular imaging, microscopic histopathology and also decreased tumor cell proliferation. Importantly, αvβ3-Fum-PD-NP effectiveness was achieved at a fractional dosage of the related clinical analogue, TNP-470. Zoledronic acid, which is commonly administered to prevent cancer induced bone fractures and to ameliorate bone pain, significantly increased bone mineralization in the Vx2 rabbit model, decreased myeloid viability in vitro and in vivo, and reduced overall tumor microvasculature. The long-acting ZA effect on the neovasculature was only a trend when compared to αvβ3-Fum-PD-NP. However, the cumulative reduction in tumor microvasculature along the tumor periphery over the study was equivalent for both treatments. Moreover, αvβ3-Fum-PD-NP and ZA further decreased tumor microvasculature additively.
In the present study, αvβ3-Fum-PD-NP was studied as an anti-angiogenic therapy in cancer. In contradistinction to anti-VEGF therapies that block the cytokine stimulation for endothelial proliferation, αvβ3-Fum-PD-NP directly targets neoendothelial cell sprouts and drives them into apoptosis. A secondary benefit of this direct anti-neoendothelial cell therapy in cancer may be a reduction in TAMs. In arthritis models, endothelial apoptosis induced by fumagillin increased intracellular NO production and local extracellular liberation that modulated synovial macrophage inflammatory responses through increased AMP-activated protein kinase (AMPK) activity. This led to decreased mammalian target of rapamycin (mTOR) activity, increased autophagic flux, increased autolysosome formation, with degradation of IkappaB kinase (IKK), suppression of the NF-κB-p65 signaling pathway and ultimately decreased inflammatory cytokine release 50. The present data obtained in the Vx2 cancer model are consistent with elements of this result. In arthritis αvβ3-Fum-PD-NP led to a near full resolution of the pathology, however in cancer, tumor viability was reduced without strong evidence of disease remission
Typically, antiangiogenic treatments, whether fumagillin or anti-VEGF, will be transient unless the cancer and macrophage cells stimulating new vessel formation are killed. Consequently, anti-angiogenic therapies are typically used as an adjuvant in combination with chemotherapy, such as 5-FU in colon cancer. In the present study, we considered the hypothesis that chronic therapy with zoledronic acid could maintain the acute transient anti-angiogenic benefits achieved with αvβ3-Fum-PD-NP, becoming an improved, potent neoadjuvant therapy. Evidence in mice and humans have suggested that ZA reduces macrophage activity, as seen with bone osteoclasts, and an anticipated pleotropic effect would be to decrease TAM stimulated angiogenesis and reduce tumor progression. ZA did decrease the microvasculature in the Vx2 tumor to similar extent as αvβ3-Fum-PD-NP but through a distinct mechanism of action. Unfortunately, the ZA reduction in vasculature, which was additive to αvβ3-Fum-PD-NP effect, did not reduce tumor volume. This result is consistent with other studies of angiogenesis inhibitors and immunotherapies that stabilize tumor progression and potentiate the anti-cancer cell effectiveness of chemotherapy.
Although ZA was effective as used clinically in strengthening the rabbit skeletal architecture, it appeared that the intracellular TAM levels of ZA achieved in vivo were inadequate to markedly reduce reactive macrophage activity and viability. Further, ZA reduced localized myeloid infiltrate and increased the anti-angiogenesis response but failed to achieve a level that would influence Vx2 tumor growth. Of note, the dramatic suppression of macrophage viability in vitro occurred at ZA dosages of 10μM and higher, otherwise the myeloid viability impact of the ZA was nonsignificant. While 10μM is achievable in the bone microenvironment in vivo, the Cmax for ZA in serum is 1μM 51. However, ZA binds to calcium hydroxyapatite present in peripheral solid tumors and bone tumors alike and may be ingested into macrophages or osteoclasts at significantly higher concentrations. In this work, ZA therapy reduced localized myeloid infiltrate and enhanced the anti-angiogenesis response, However, an effective myeloid intracellular IC50 dose to substantively complement the anti-tumor αvβ3-Fum-PD NP response was not achieved. The intracellular concentrations of the compound by phagocytosis of microcalcifications maybe too low to induce robust macrophage inhibition even though the microcalcification density observed in rabbits was far higher than that observed in the human breast cancer specimens. Numerous but variable observations of ZA benefit in patients suggests that the drug in combination with αvβ3-Fum-PD-NP has significant neoadjuvant potential if the delivery of ZA into myeloid cells is increased. This concept was illustrated in xenograft prostate cancer mouse models that received IV ZA, ZA PEGylated nanoparticles (ZA-PEG NPs) or ZA PEGylated liposomes (LIPO-ZA). Both particle systems relied on nonspecific macrophage uptake. These ZA delivery platforms offered superior anti-cancer effects when compared to intravenous ZA, which had no benefit in this mouse cancer model. The best treatment results were obtained with ZA-PEG NPs, which reduced TAM numbers in tumor xenografts, decreased tumor vascularity and increased cancer necrosis and apoptosis 52, 53. Perhaps a dual therapy, single nanoplatform providing αvβ3-targeting of Fum-PD and ZA would provide the potent neoadjuvant benefits sought in the present study.
Supplementary Material
Acknowledgments
Financial support: This research was supported in whole or part by grants from the NIH CA154737 (KNW/DP/GML); CA097250 (KNW); AR067491 (SAW), DK102691 (SAW), and HL073646 (SAW), HL122471 (GML), HL112518 (GML), HL113392 (GML), HHSN26820140042C (GML) and training grants 5T32CA113275-07 (AKE) and 5T32GM007067-39 (MHR). We appreciate the further support provided by the Washington University Musculoskeletal Research Center (NIH P30AR057235), Washington University Digestive Diseases Research Core (NIH P30DK52574), Hope Center Alafi Neuroimaging Lab (S10RR027552), The Barnes-Jewish Research Foundation and The St. Louis Men’s Group Against Cancer.
Footnotes
Conflict of Interest: None.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer. 2008;8:592–603. doi: 10.1038/nrc2442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bruno A, Pagani A, Pulze L, Albini A, Dallaglio K, Noonan DM, et al. Orchestration of angiogenesis by immune cells. Front Oncol. 2014;4:131. doi: 10.3389/fonc.2014.00131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74. doi: 10.1016/j.cell.2011.02.013. [DOI] [PubMed] [Google Scholar]
- 4.Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer. 2008;8:618–31. doi: 10.1038/nrc2444. [DOI] [PubMed] [Google Scholar]
- 5.Ingber D, Fujita T, Kishimoto S, Sudo K, Kanamaru T, Brem H, et al. Synthetic analogues of fumagillin that inhibit angiogenesis and suppress tumour growth. Nature. 1990;348:555–7. doi: 10.1038/348555a0. [DOI] [PubMed] [Google Scholar]
- 6.Sin N, Meng L, Wang MQ, Wen JJ, Bornmann WG, Crews CM. The anti-angiogenic agent fumagillin covalently binds and inhibits the methionine aminopeptidase, MetAP-2. Proc Natl Acad Sci U S A. 1997;94:6099–103. doi: 10.1073/pnas.94.12.6099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Liu S, Widom J, Kemp CW, Crews CM, Clardy J. Structure of human methionine aminopeptidase-2 complexed with fumagillin. Science. 1998;282:1324–7. doi: 10.1126/science.282.5392.1324. [DOI] [PubMed] [Google Scholar]
- 8.Lowther WT, McMillen DA, Orville AM, Matthews BW. The anti-angiogenic agent fumagillin covalently modifies a conserved active-site histidine in the Escherichia coli methionine aminopeptidase. Proc Natl Acad Sci U S A. 1998;95:12153–7. doi: 10.1073/pnas.95.21.12153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Turk BE, Griffith EC, Wolf S, Biemann K, Chang YH, Liu JO. Selective inhibition of amino-terminal methionine processing by TNP-470 and ovalicin in endothelial cells. Chem Biol. 1999;6:823–33. doi: 10.1016/s1074-5521(99)80129-x. [DOI] [PubMed] [Google Scholar]
- 10.Wang J, Lou P, Henkin J. Selective inhibition of endothelial cell proliferation by fumagillin is not due to differential expression of methionine aminopeptidases. J Cell Biochem. 2000;77:465–73. [PubMed] [Google Scholar]
- 11.Castronovo V, Belotti D. TNP-470 (AGM-1470): mechanisms of action and early clinical development. Eur J Cancer. 1996;32A:2520–7. doi: 10.1016/s0959-8049(96)00388-7. [DOI] [PubMed] [Google Scholar]
- 12.Konno H, Tanaka T, Kanai T, Maruyama K, Nakamura S, Baba S. Efficacy of an angiogenesis inhibitor, TNP-470, in xenotransplanted human colorectal cancer with high metastatic potential. Cancer. 1996;77:1736–40. doi: 10.1002/(SICI)1097-0142(19960415)77:8<1736::AID-CNCR48>3.0.CO;2-Y. [DOI] [PubMed] [Google Scholar]
- 13.Shusterman S, Grupp SA, Barr R, Carpentieri D, Zhao H, Maris JM. The angiogenesis inhibitor TNP-470 effectively inhibits human neuroblastoma xenograft growth, especially in the setting of subclinical disease. Clin Cancer Res. 2001;7:977–84. [PubMed] [Google Scholar]
- 14.Kudelka AP, Verschraegen CF, Loyer E. Complete remission of metastatic cervical cancer with the angiogenesis inhibitor TNP-470. N Engl J Med. 1998;338:991–2. doi: 10.1056/NEJM199804023381412. [DOI] [PubMed] [Google Scholar]
- 15.Bhargava P, Marshall JL, Rizvi N, Dahut W, Yoe J, Figuera M, et al. A Phase I and pharmacokinetic study of TNP-470 administered weekly to patients with advanced cancer. Clin Cancer Res. 1999;5:1989–95. [PubMed] [Google Scholar]
- 16.Logothetis CJ, Wu KK, Finn LD, Daliani D, Figg W, Ghaddar H, et al. Phase I trial of the angiogenesis inhibitor TNP-470 for progressive androgen-independent prostate cancer. Clin Cancer Res. 2001;7:1198–203. [PubMed] [Google Scholar]
- 17.Winter P, Neubauer A, Caruthers S, Harris T, Robertson J, Williams T, et al. Endothelial alpha(nu)beta(3)-Integrin targeted fumagillin nanoparticles inhibit angiogenesis in atherosclerosis. Arterioscler Thromb Vasc Biol. 2006;26:2103–2109. doi: 10.1161/01.ATV.0000235724.11299.76. [DOI] [PubMed] [Google Scholar]
- 18.Winter PM, Schmieder AH, Caruthers SD, Keene JL, Zhang H, Wickline SA, et al. Minute dosages of alpha(nu)beta3-targeted fumagillin nanoparticles impair Vx-2 tumor angiogenesis and development in rabbits. FASEB J. 2008;22:2758–67. doi: 10.1096/fj.07-103929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Schmieder AH, Caruthers SD, Zhang H, Williams TA, Robertson JD, Wickline SA, et al. Three-dimensional MR mapping of angiogenesis with {alpha}5{beta}1({alpha}{nu}{beta}3)-targeted theranostic nanoparticles in the MDA-MB-435 xenograft mouse model. FASEB J. 2008;22:4179–4189. doi: 10.1096/fj.08-112060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lanza GM, Yu X, Winter PM, Abendschein DR, Karukstis KK, Scott MJ, et al. Targeted antiproliferative drug delivery to vascular smooth muscle cells with a magnetic resonance imaging nanoparticle contrast agent: implications for rational therapy of restenosis. Circulation. 2002;106:2842–7. doi: 10.1161/01.cir.0000044020.27990.32. [DOI] [PubMed] [Google Scholar]
- 21.Partlow K, Lanza G, Wickline S. Exploiting lipid raft transport with membrane targeted nanoparticles: A strategy for cytosolic drug delivery. Biomaterials. 2008;29:3367–3375. doi: 10.1016/j.biomaterials.2008.04.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Soman N, Lanza G, Heuser J, Schlesinger P, Wickline S. Synthesis and characterization of stable fluorocarbon nanostructures as drug delivery vehicles for cytolytic peptides. Nano Lett. 2008;8:1131–1136. doi: 10.1021/nl073290r. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Pan D, Sanyal N, Schmieder AH, Senpan A, Kim B, Yang X, et al. Antiangiogenic nanotherapy with lipase-labile Sn-2 fumagillin prodrug. Nanomedicine (Lond) 2012;7:1507–19. doi: 10.2217/nnm.12.27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Zhang R, Pan D, Cai X, Yang X, Senpan A, Allen JS, et al. Alphanubeta(3)-targeted copper nanoparticles incorporating an Sn 2 lipase-labile fumagillin prodrug for photoacoustic neovascular imaging and treatment. Theranostics. 2015;5:124–33. doi: 10.7150/thno.10014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zhou HF, Yan H, Senpan A, Wickline SA, Pan D, Lanza GM, et al. Suppression of inflammation in a mouse model of rheumatoid arthritis using targeted lipase-labile fumagillin prodrug nanoparticles. Biomaterials. 2012;33:8632–40. doi: 10.1016/j.biomaterials.2012.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Hirbe AC, Roelofs AJ, Floyd DH, Deng H, Becker SN, Lanigan LG, et al. The bisphosphonate zoledronic acid decreases tumor growth in bone in mice with defective osteoclasts. Bone. 2009;44:908–16. doi: 10.1016/j.bone.2009.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Aft R, Naughton M, Trinkaus K, Watson M, Ylagan L, Chavez-MacGregor M, et al. Effect of zoledronic acid on disseminated tumour cells in women with locally advanced breast cancer: an open label, randomised, Phase 2 trial. Lancet Oncol. 2010;11:421–8. doi: 10.1016/S1470-2045(10)70054-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Clezardin P. Bisphosphonates’ antitumor activity: an unravelled side of a multifaceted drug class. Bone. 2011;48:71–9. doi: 10.1016/j.bone.2010.07.016. [DOI] [PubMed] [Google Scholar]
- 29.Coleman R, Gnant M, Morgan G, Clezardin P. Effects of bone-targeted agents on cancer progression and mortality. J Natl Cancer Inst. 2012;104:1059–67. doi: 10.1093/jnci/djs263. [DOI] [PubMed] [Google Scholar]
- 30.Rogers TL, Wind N, Hughes R, Nutter F, Brown HK, Vasiliadou I, et al. Macrophages as potential targets for zoledronic acid outside the skeleton-evidence from in vitro and in vivo models. Cell Oncol (Dordr) 2013;36:505–14. doi: 10.1007/s13402-013-0156-2. [DOI] [PubMed] [Google Scholar]
- 31.Mathew A, Brufsky A. Breast cancer: Zoledronic acid--more than just a bone drug. Nature reviews Clin Oncol. 2014;11:564–5. doi: 10.1038/nrclinonc.2014.152. [DOI] [PubMed] [Google Scholar]
- 32.Worsley DF, Lentle BC. Uptake of technetium-99m MDP in primary amyloidosis with a review of the mechanisms of soft tissue localization of bone seeking radiopharmaceuticals. J Nuclear Med. 1993;34:1612–5. [PubMed] [Google Scholar]
- 33.Junankar S, Shay G, Jurczyluk J, Ali N, Down J, Pocock N, et al. Real-time intravital imaging establishes tumor-associated macrophages as the extraskeletal target of bisphosphonate action in cancer. Cancer Discov. 2015;5:35–42. doi: 10.1158/2159-8290.CD-14-0621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Heller E, Hurchla MA, Xiang J, Su X, Chen S, Schneider J, et al. Hedgehog signaling inhibition blocks growth of resistant tumors through effects on tumor microenvironment. Cancer Res. 2012;72:897–907. doi: 10.1158/0008-5472.CAN-11-2681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Amend SR, Uluckan O, Hurchla M, Leib D, Novack DV, Silva M, et al. Thrombospondin-1 regulates bone homeostasis through effects on bone matrix integrity and nitric oxide signaling in osteoclasts. J Bone Miner Res. 2015;30:106–15. doi: 10.1002/jbmr.2308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Su X, Floyd DH, Hughes A, Xiang J, Schneider JG, Uluckan O, et al. The ADP receptor P2RY12 regulates osteoclast function and pathologic bone remodeling. J Clin Invest. 2012;122:3579–92. doi: 10.1172/JCI38576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Pan D, Schmieder AH, Wang K, Yang X, Senpan A, Cui G, et al. Anti-angiogenesis therapy in the Vx2 rabbit cancer model with a lipase-cleavable Sn 2 taxane phospholipid prodrug using alpha(v)beta(3)-targeted theranostic nanoparticles. Theranostics. 2014;4:565–78. doi: 10.7150/thno.7581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Pan D, Pramanik M, Senpan A, Allen JS, Zhang H, Wickline SA, et al. Molecular photoacoustic imaging of angiogenesis with integrin-targeted gold nanobeacons. FASEB J. 2011;25:875–82. doi: 10.1096/fj.10-171728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wang K, Pan D, Schmieder AH, Senpan A, Hourcade DE, Pham CT, et al. Synergy between surface and core entrapped metals in a mixed manganese-gadolinium nanocolloid affords safer MR imaging of sparse biomarkers. Nanomedicine. 2015;11:601–9. doi: 10.1016/j.nano.2014.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Wang K, Pan D, Schmieder AH, Senpan A, Caruthers SD, Cui G, et al. Atherosclerotic neovasculature MR imaging with mixed manganese-gadolinium nanocolloids in hyperlipidemic rabbits. Nanomedicine. 2015;11:569–78. doi: 10.1016/j.nano.2014.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Uluckan O, Becker SN, Deng H, Zou W, Prior JL, Piwnica-Worms D, et al. CD47 regulates bone mass and tumor metastasis to bone. Cancer Res. 2009;69:3196–204. doi: 10.1158/0008-5472.CAN-08-3358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bouxsein ML, Boyd SK, Christiansen BA, Guldberg RE, Jepsen KJ, Muller R. Guidelines for assessment of bone microstructure in rodents using micro-computed tomography. J Bone Mine Res. 2010;25:1468–86. doi: 10.1002/jbmr.141. [DOI] [PubMed] [Google Scholar]
- 43.Schmieder AH, Winter PM, Williams TA, Allen JS, Hu G, Zhang H, et al. Molecular MR imaging of neovascular progression in the Vx2 tumor with alpha v beta 3-targeted paramagnetic nanoparticles. Radiology. 2013;268:470–80. doi: 10.1148/radiol.13120789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Herbst RS, Madden TL, Tran HT, Blumenschein GR, Jr, Meyers CA, Seabrooke LF, et al. Safety and pharmacokinetic effects of TNP-470, an angiogenesis inhibitor, combined with paclitaxel in patients with solid tumors: evidence for activity in non-small-cell lung cancer. J Clin Oncol. 2002;20:4440–7. doi: 10.1200/JCO.2002.04.006. [DOI] [PubMed] [Google Scholar]
- 45.Dezube BJ, Von Roenn JH, Holden-Wiltse J, Cheung TW, Remick SC, Cooley TP, et al. Fumagillin analog in the treatment of Kaposi’s sarcoma: a Phase I AIDS Clinical Trial Group study. AIDS Clinical Trial Group No. 215 Team. J Clin Oncol. 1998;16:1444–9. doi: 10.1200/JCO.1998.16.4.1444. [DOI] [PubMed] [Google Scholar]
- 46.Winter PM, Schmieder AH, Caruthers SD, Keene JL, Zhang H, Wickline SA, et al. Minute dosages of alpha(nu)beta3-targeted fumagillin nanoparticles impair Vx-2 tumor angiogenesis and development in rabbits. FASEB J. 2008;22:2758–2767. doi: 10.1096/fj.07-103929. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Dirkx AE, Oude Egbrink MG, Kuijpers MJ, van der Niet ST, Heijnen VV, Boumater Steege JC, et al. Tumor angiogenesis modulates leukocyte-vessel wall interactions in vivo by reducing endothelial adhesion molecule expression. Cancer Res. 2003;63:2322–2329. [PubMed] [Google Scholar]
- 48.Griffioen AW, Damen CA, Mayo KH, Barendsz-Janson AF, Martinotti S, Blijham GH, et al. Angiogenesis inhibitors overcome tumor induced endothelial cell anergy. Int J Cancer. 1999;80:315–319. doi: 10.1002/(sici)1097-0215(19990118)80:2<315::aid-ijc23>3.0.co;2-l. [DOI] [PubMed] [Google Scholar]
- 49.Dirkx AE, oude Egbrink MG, Castermans K, van der Schaft DW, Thijssen VL, Dings RP, et al. Anti-angiogenesis therapy can overcome endothelial cell anergy and promote leukocyte-endothelium interactions and infiltration in tumors. FASEB J. 2006;20:621–630. doi: 10.1096/fj.05-4493com. [DOI] [PubMed] [Google Scholar]
- 50.Zhou H, Yan H, Hu Y, Springer L, Yang X, Wickline S, et al. Fumagillin prodrug nanotherapy suppresses macrophage inflammatory response via endothelial nitric oxide. ACS Nano. 2014;8:7305–7317. doi: 10.1021/nn502372n. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sato M, Grasser W, Endo N, Akins R, Simmons H, Thompson DD, et al. Bisphosphonate action. Alendronate localization in rat bone and effects on osteoclast ultrastructure. J Clin Invest. 1991;88:2095–2105. doi: 10.1172/JCI115539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Marra M, Salzano G, Leonetti C, Porru M, Franco R, Zappavigna S, et al. New self-assembly nanoparticles and stealth liposomes for the delivery of zoledronic acid: a comparative study. Biotechnol Adv. 2012;30:302–9. doi: 10.1016/j.biotechadv.2011.06.018. [DOI] [PubMed] [Google Scholar]
- 53.Marra M, Abbruzzese A, Addeo R, Del Prete S, Tassone P, Tonini G, et al. Cutting the limits of aminobisphosphonates: new strategies for the potentiation of their anti-tumour effects. Curr Cancer Drug Targets. 2009;9:791–800. doi: 10.2174/156800909789760285. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.







