Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 546
1.
Cancer Med ; 12(18): 18981-18987, 2023 09.
Article En | MEDLINE | ID: mdl-37680049

BACKGROUND: Tumor-associated angiogenesis mediates the growth and metastasis of most solid cancers. Targeted therapies of the VEGF pathways can effectively block these processes but often fail to provide lasting benefits due to acquired resistance and complications. RESULTS: Recently, we discovered ßIV -spectrin as a powerful regulator of angiogenesis and potential new target. We previously reported that ßIV -spectrin is dynamically expressed in endothelial cells (EC) to induce VEGFR2 protein turnover during development. Here, we explored how ßIV -spectrin influences the tumor vasculature using the murine B16 melanoma model and determined that loss of EC-specific ßIV -spectrin dramatically promotes tumor growth and metastasis. Intraperitoneally injected B16 cells formed larger tumors with increased tumor vessel density and greater propensity for metastatic spread particularly to the chest cavity and lung compared to control mice. These results support ßIV -spectrin as a key regulator of tumor angiogenesis and a viable vascular target in cancer.


Melanoma, Experimental , Spectrin , Animals , Mice , Endothelial Cells/metabolism , Melanoma, Experimental/blood supply , Neovascularization, Pathologic , Spectrin/metabolism
2.
Am J Pathol ; 192(2): 379-388, 2022 02.
Article En | MEDLINE | ID: mdl-34861214

Vascular endothelial growth factor (VEGF) blockers are used widely in clinics to target various types of human cancer. Although VEGF blockers exert marked tumor suppressive effects, the therapeutic effects can be limited. Moreover, accumulating evidence shows that VEGF acts not just on endothelial cells but also on various nonendothelial cells, including tumor and immune cells, suggesting a need to revisit the bona fide action of VEGF on endothelial cells using specific genetic mouse models. Herein, tamoxifen-inducible endothelial-specific knockout mice lacking VEGF receptor 2 (Vegfr2), the major signal transducer for VEGF, were used. The initial event resulting from cessation of endothelial Vegfr2 signaling was vascular truncation and fragmentation, rather than maturation of abnormalized vessels. Although deletion of endothelial Vegfr2 suppressed intratumor hemorrhage, it enhanced hypoxia in tumor cells and reduced the number of infiltrating cytotoxic T cells, suggesting a profound reduction in intratumor blood flow. In various tissues, deletion of endothelial Vegfr2 induced regression of healthy capillaries in intestinal villi, substantiating intestinal perforation, which is one of the most common adverse effects of VEGF blockade in humans. Overall, the data suggest that some of the known effects of VEGF blockers on tumor vessels are caused by partial cessation of VEGF signaling, or by actions on nonendothelial cells. The results increase the understanding of the mechanisms underlying anti-angiogenic therapy.


Endothelial Cells , Gene Deletion , Melanoma, Experimental , Neoplasm Proteins , Neovascularization, Pathologic , Vascular Endothelial Growth Factor Receptor-2 , Animals , Cell Hypoxia/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Melanoma, Experimental/blood supply , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
3.
BMB Rep ; 54(9): 470-475, 2021 Sep.
Article En | MEDLINE | ID: mdl-34488932

Low-dose metronomic chemotherapy has been introduced as a less toxic and effective strategy to inhibit tumor angiogenesis, but its anti-angiogenic mechanism on endothelial progenitor cells (EPCs) has not been fully elucidated. Here, we investigated the functional role of regulated in development and DNA damage response 1 (REDD1), an endogenous inhibitor of mTORC1, in low-dose doxorubicin (DOX)-mediated dysregulation of EPC functions. DOX treatment induced REDD1 expression in bone marrow mononuclear cells (BMMNCs) and subsequently reduced mTORC1-dependent translation of endothelial growth factor (VEGF) receptor (Vegfr)-2 mRNA, but not that of the mRNA transcripts for Vegfr-1, epidermal growth factor receptor, and insulin-like growth factor-1 receptor. This selective event was a risk factor for the inhibition of BMMNC differentiation into EPCs and their angiogenic responses to VEGF-A, but was not observed in Redd1-deficient BMMNCs. Low-dose metronomic DOX treatment reduced the mobilization of circulating EPCs in B16 melanoma-bearing wild-type but not Redd1-deficient mice. However, REDD1 overexpression inhibited the differentiation and mobilization of EPCs in both wild-type and Redd1-deficient mice. These data suggest that REDD1 is crucial for metronomic DOX-mediated EPC dysfunction through the translational repression of Vegfr-2 transcript, providing REDD1 as a potential therapeutic target for the inhibition of tumor angiogenesis and tumor progression. [BMB Reports 2021; 54(9): 470-475].


Cell Differentiation/drug effects , Down-Regulation/drug effects , Doxorubicin/pharmacology , Transcription Factors/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Cultured , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Mice, Knockout , Neovascularization, Pathologic , Nitric Oxide/metabolism , Transcription Factors/deficiency , Transcription Factors/genetics , Transplantation, Heterologous , Vascular Endothelial Growth Factor A/pharmacology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics
4.
Microvasc Res ; 138: 104209, 2021 11.
Article En | MEDLINE | ID: mdl-34146582

Angiogenesis is critical for many diseases. Previously, we reported that Down Syndrome Candidate Region 1 isoform 1L (DSCR1-1L) was one of the most up-regulated genes in endothelial cells induced by VEGF and histamine, and regulated endothelial cell proliferation and Matrigel angiogenesis in mice. However, it was not known whether DSCR1-1L regulated angiogenesis in vivo and what was the molecular mechanism underlying it. In this study, gene knockdown and overexpression models were established to study the role of DSCR1-1L in angiogenesis in vivo. Further, the downstream regulatory target of DSCR1-1L was explored with molecular biological methods in vascular endothelial cells. We found that DSCR1-1L shRNAs significantly inhibited angiogenesis induced by VEGF in mice (p < 0.0001). In the gain-of-function assay, overexpression of DSCR1-1L cDNA in mouse endothelium of EC-FH-DSCR1-1L transgenic mice was sufficient to induce angiogenesis significantly (p < 0.01). DSCR1-1L regulated angiogenesis in the early stage by down-regulation of the VE-cadherin expression through targeting its transcription, but not mRNA stability. Three DSCR1-1L-targeted DNA elements in the VE-cadherin promoter were identified by promoter reporter assays, among which, a novel specific transcriptional complex was found. The DNA sequence (CTTCTG) in the VE-cadherin promoter was identified to directly interact with proteins by Electrophoresis Mobility Shift Assays and DNase I footprint assay. Hence, DSCR1-1L is an excellent therapeutic target for angiogenic diseases through down-regulating the formation of a novel transcriptional complex on the VE-cadherin promoter. DSCR1-1L shRNAs and cDNA have the potential to be developed for clinical application. Our results also contribute significantly to the field of mechanistic studies.


Antigens, CD/metabolism , Cadherins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Melanoma, Experimental/blood supply , Muscle Proteins/metabolism , Neovascularization, Pathologic , Neovascularization, Physiologic , Promoter Regions, Genetic , Animals , Antigens, CD/genetics , Cadherins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice, Nude , Mice, Transgenic , Muscle Proteins/genetics , Signal Transduction
5.
Appl Radiat Isot ; 174: 109778, 2021 Aug.
Article En | MEDLINE | ID: mdl-34004593

Formation and growth of metastases require a new vascular network. Angiogenesis plays an essential role in the expansion and progression of most malignancies. A high number of molecular pathways regulate angiogenesis, including vascular endothelial growth factor (VEGF), αvß3 integrin, matrix metalloproteinases (MMPs), or aminopeptidase N. The aim of this study is to involve new, easily accessible peptide sequences into the of neo-angiogenesis in malignant processes. Labelling of these peptide ligands with 68Ga enable PET imaging of neo-vascularization.


Gallium Radioisotopes/chemistry , Melanoma, Experimental/blood supply , Neovascularization, Pathologic/diagnostic imaging , Peptides/chemistry , Positron-Emission Tomography/methods , Animals , CD13 Antigens/metabolism , Chromatography, High Pressure Liquid/methods , Gallium Radioisotopes/pharmacokinetics , Integrin alphaVbeta3/metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Tissue Distribution , Vascular Endothelial Growth Factor Receptor-1/metabolism
6.
Angiogenesis ; 24(3): 597-611, 2021 08.
Article En | MEDLINE | ID: mdl-33629198

Antibiotic-induced microbial imbalance, or dysbiosis, has systemic and long-lasting effects on the host and response to cancer therapies. However, the effects on tumor endothelial cells are largely unknown. Therefore, the goal of the current study was to generate matched B16-F10 melanoma associated endothelial cell lines isolated from mice with and without antibiotic-induced dysbiosis. After validating endothelial cell markers on a genomic and proteomic level, functional angiogenesis assays (i.e., migration and tube formation) also confirmed their vasculature origin. Subsequently, we found that tumor endothelial cells derived from dysbiotic mice (TEC-Dys) were more sensitive to ionizing radiotherapy in the range of clinically-relevant hypofractionated doses, as compared to tumor endothelial cells derived from orthobiotic mice (TEC-Ortho). In order to identify tumor vasculature-associated drug targets during dysbiosis, we used tandem mass tag mass spectroscopy and focused on the statistically significant cellular membrane proteins overexpressed in TEC-Dys. By these criteria c-Met was the most differentially expressed protein, which was validated histologically by comparing tumors with or without dysbiosis. Moreover, in vitro, c-Met inhibitors Foretinib, Crizotinib and Cabozantinib were significantly more effective against TEC-Dys than TEC-Ortho. In vivo, Foretinib inhibited tumor growth to a greater extent during dysbiosis as compared to orthobiotic conditions. Thus, we surmise that tumor response in dysbiotic patients may be greatly improved by targeting dysbiosis-induced pathways, such as c-Met, distinct from the many targets suppressed due to dysbiosis.


Dysbiosis , Endothelial Cells/enzymology , Melanoma, Experimental , Neovascularization, Pathologic , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met , Animals , Dysbiosis/enzymology , Dysbiosis/microbiology , Melanoma, Experimental/blood supply , Melanoma, Experimental/enzymology , Melanoma, Experimental/microbiology , Melanoma, Experimental/therapy , Mice , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/microbiology , Neovascularization, Pathologic/therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Radiotherapy
7.
J Exp Clin Cancer Res ; 39(1): 266, 2020 Nov 30.
Article En | MEDLINE | ID: mdl-33256804

BACKGROUND: Tumor angiogenesis is regarded as a rational anti-cancer target. The efficacy and indications of anti-angiogenic therapies in clinical practice, however, are relatively limited. Therefore, there still exists a demand for revealing the distinct characteristics of tumor endothelium that is crucial for the pathological angiogenesis. L-type amino acid transporter 1 (LAT1) is well known to be highly and broadly upregulated in tumor cells to support their growth and proliferation. In this study, we aimed to establish the upregulation of LAT1 as a novel general characteristic of tumor-associated endothelial cells as well, and to explore the functional relevance in tumor angiogenesis. METHODS: Expression of LAT1 in tumor-associated endothelial cells was immunohistologically investigated in human pancreatic ductal adenocarcinoma (PDA) and xenograft- and syngeneic mouse tumor models. The effects of pharmacological and genetic ablation of endothelial LAT1 were examined in aortic ring assay, Matrigel plug assay, and mouse tumor models. The effects of LAT1 inhibitors and gene knockdown on cell proliferation, regulation of translation, as well as on the VEGF-A-dependent angiogenic processes and intracellular signaling were investigated in in vitro by using human umbilical vein endothelial cells. RESULTS: LAT1 was highly expressed in vascular endothelial cells of human PDA but not in normal pancreas. Similarly, high endothelial LAT1 expression was observed in mouse tumor models. The angiogenesis in ex/in vivo assays was suppressed by abrogating the function or expression of LAT1. Tumor growth in mice was significantly impaired through the inhibition of angiogenesis by targeting endothelial LAT1. LAT1-mediated amino acid transport was fundamental to support endothelial cell proliferation and translation initiation in vitro. Furthermore, LAT1 was required for the VEGF-A-dependent migration, invasion, tube formation, and activation of mTORC1, suggesting a novel cross-talk between pro-angiogenic signaling and nutrient-sensing in endothelial cells. CONCLUSIONS: These results demonstrate that the endothelial LAT1 is a novel key player in tumor angiogenesis, which regulates proliferation, translation, and pro-angiogenic VEGF-A signaling. This study furthermore indicates a new insight into the dual functioning of LAT1 in tumor progression both in tumor cells and stromal endothelium. Therapeutic inhibition of LAT1 may offer an ideal option to potentiate anti-angiogenic therapies.


Amino Acid Transport Systems/metabolism , Carcinoma, Pancreatic Ductal/blood supply , Endothelium, Vascular/metabolism , Large Neutral Amino Acid-Transporter 1/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Pancreatic Neoplasms/blood supply , Vascular Endothelial Growth Factor A/metabolism , Amino Acid Transport System y+L/metabolism , Animals , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Endothelium, Vascular/pathology , Female , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Melanoma, Experimental/blood supply , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/metabolism , Signal Transduction
8.
Cells ; 9(11)2020 11 13.
Article En | MEDLINE | ID: mdl-33202705

The role of tumor necrosis factor-α (TNF-α) in shaping the tumor microenvironment is ambiguous. Consistent with its uncertain role in melanoma, TNF-α plays a dual role, either acting as a cytotoxic cytokine or favoring a tumorigenic inflammatory microenvironment. TNF-α signals via two cognate receptors, namely TNFR1 (p55) and TNFR2 (p75), which mediate divergent biological activities. Here, we analyzed the impact of TNFR1 deficiency in tumor progression in the B16.F1 melanoma model. Tumors developed in mice lacking TNFR1 (TNFR1 knock-out; KO) were smaller and displayed lower proliferation compared to their wild type (WT) counterpart. Moreover, TNFR1 KO mice showed reduced tumor angiogenesis. Although no evidence of spontaneous metastases was observed, conditioned media obtained from TNFR1 KO tumors increased tumor cell migration. Whereas the analysis of tumor-associated immune cell infiltrates showed similar frequency of total and M2-polarized tumor-associated macrophages (TAMs), the percentage of CD8+ T cells was augmented in TNFR1 KO tumors. Indeed, functional ex vivo assays demonstrated that CD8+ T cells obtained from TNFR1KO mice displayed an increased cytotoxic function. Thus, lack of TNFR1 attenuates melanoma growth by modulating tumor cell proliferation, migration, angiogenesis and CD8+ T cell accumulation and activation, suggesting that interruption of TNF-TNFR1 signaling may contribute to control tumor burden.


CD8-Positive T-Lymphocytes/immunology , Melanoma, Experimental/blood supply , Melanoma, Experimental/immunology , Neovascularization, Pathologic/immunology , Receptors, Tumor Necrosis Factor, Type I/deficiency , Animals , Cell Proliferation , Lymphocyte Activation/immunology , Melanins/metabolism , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , Tumor Microenvironment/immunology
9.
Cancer Lett ; 495: 66-75, 2020 12 28.
Article En | MEDLINE | ID: mdl-32891714

Melanoma, a form of skin cancer, is one of the most common cancers in young men and women. Tumors require angiogenesis to provide oxygen and nutrients for growth. Pro-angiogenic molecules such as VEGF and anti-angiogenic molecules such as sFlt-1 control angiogenesis. In addition, the serum protein, Beta2 Glycoprotein I (ß2-GPI) induces or inhibits angiogenesis depending on conformation and concentration. ß2-GPI binds to proteins and negatively charged phospholipids on hypoxic endothelial cells present in the tumor microenvironment. We hypothesized that peptides derived from the binding domain of ß2-GPI would regulate angiogenesis and melanoma growth. In vitro analyses determined the peptides reduced endothelial cell migration and sFlt-1 secretion. In a syngeneic, immunocompetent mouse melanoma model, ß2-GPI-derived peptides also reduced melanoma growth in a dose-dependent response with increased sFlt-1 and attenuated vascular markers compared to negative controls. Importantly, administration of peptide with sFlt-1 antibody resulted in tumor growth. These data demonstrate the therapeutic potential of novel ß2-GPI-derived peptides to attenuate tumor growth and endothelial migration is sFlt-1 dependent.


Endothelial Cells/cytology , Melanoma, Experimental/drug therapy , Peptides/administration & dosage , Vascular Endothelial Growth Factor Receptor-1/metabolism , beta 2-Glycoprotein I/chemistry , Animals , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Female , Melanoma, Experimental/blood supply , Melanoma, Experimental/metabolism , Mice , Peptides/pharmacology , Phospholipids/metabolism , Protein Domains , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays , beta 2-Glycoprotein I/metabolism
10.
Oncogene ; 39(41): 6480-6492, 2020 10.
Article En | MEDLINE | ID: mdl-32879446

Angiogenesis inhibitors, such as the receptor tyrosine kinase (RTK) inhibitor sunitinib, target vascular endothelial growth factor (VEGF) signaling in cancers. However, only a fraction of patients respond, and most ultimately develop resistance to current angiogenesis inhibitor therapies. Activity of alternative pro-angiogenic growth factors, acting via RTK or G-protein coupled receptors (GPCR), may mediate VEGF inhibitor resistance. The phosphoinositide 3-kinase (PI3K)ß isoform is uniquely coupled to both RTK and GPCRs. We investigated the role of endothelial cell (EC) PI3Kß in tumor angiogenesis. Pro-angiogenic GPCR ligands were expressed by patient-derived renal cell carcinomas (PD-RCC), and selective inactivation of PI3Kß reduced PD-RCC-stimulated EC spheroid sprouting. EC-specific PI3Kß knockout (ΕC-ßKO) in mice potentiated the sunitinib-induced reduction in subcutaneous growth of LLC1 and B16F10, and lung metastasis of B16F10 tumors. Compared to single-agent sunitinib treatment, tumors in sunitinib-treated ΕC-ßKO mice showed a marked decrease in microvessel density, and reduced new vessel formation. The fraction of perfused mature tumor microvessels was increased in ΕC-ßKO mice suggesting immature microvessels were most sensitive to combined sunitinib and PI3Kß inactivation. Taken together, EC PI3Kß inactivation with sunitinib inhibition reduces microvessel turnover and decreases heterogeneity of the tumor microenvironment, hence PI3Kß inhibition may be a useful adjuvant antiangiogenesis therapy with sunitinib.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Renal Cell/pathology , Class I Phosphatidylinositol 3-Kinases/metabolism , Kidney Neoplasms/pathology , Neovascularization, Pathologic/pathology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/blood supply , Carcinoma, Renal Cell/drug therapy , Cell Line, Tumor , Cell Proliferation/drug effects , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , Endothelium, Vascular/cytology , Endothelium, Vascular/pathology , Human Umbilical Vein Endothelial Cells , Humans , Kidney Neoplasms/blood supply , Kidney Neoplasms/drug therapy , Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice, Knockout , Microvessels/drug effects , Microvessels/pathology , Morpholines/pharmacology , Morpholines/therapeutic use , Neovascularization, Pathologic/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Sunitinib/pharmacology , Sunitinib/therapeutic use , Thiazoles/pharmacology , Thiazoles/therapeutic use , Tumor Microenvironment/drug effects , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
11.
Sci Rep ; 10(1): 13245, 2020 08 06.
Article En | MEDLINE | ID: mdl-32764623

Despite advances in therapy for melanoma, heterogeneous responses with limited durability represent a major gap in treatment outcomes. The purpose of this study was to determine whether alteration in tumor blood flow could augment drug delivery and improve antitumor responses in a regional model of melanoma. This approach to altering tumor blood flow was termed "dynamic control." Dynamic control of tumor vessels in C57BL/6 mice bearing B16 melanoma was performed using volume expansion (saline bolus) followed by phenylephrine. Intravital microscopy (IVM) was used to observe changes directly in real time. Our approach restored blood flow in non-functional tumor vessels. It also resulted in increased chemotherapy (melphalan) activity, as measured by formation of DNA adducts. The combination of dynamic control and melphalan resulted in superior outcomes compared to melphalan alone (median time to event 40.0 vs 25.0 days, respectively, p = 0.041). Moreover, 25% (3/12) of the mice treated with the combination approach showed complete tumor response. Importantly, dynamic control plus melphalan did not result in increased adverse events. In summary, we showed that dynamic control was feasible, directly observable, and augmented antitumor responses in a regional model of melanoma. Early clinical trials to determine the translational feasibility of dynamic control are ongoing.


Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Melphalan/administration & dosage , Phenylephrine/administration & dosage , Saline Solution/administration & dosage , Animals , Blood Circulation/drug effects , Cell Line, Tumor , Drug Synergism , Feasibility Studies , Female , Intravital Microscopy , Melphalan/pharmacology , Mice , Mice, Inbred C57BL , Phenylephrine/pharmacology , Saline Solution/pharmacology , Survival Analysis , Treatment Outcome , Xenograft Model Antitumor Assays
12.
Biomater Sci ; 8(11): 3052-3062, 2020 Jun 07.
Article En | MEDLINE | ID: mdl-32347852

Inefficient nanoparticle accumulation in solid tumors hinders the clinical translation of cancer nanomedicines. Herein, we proposed that sildenafil, a vasodilator ampholyte, could be used to promote nanoparticle accumulation by inducing vasodilation after its tumor acidity-triggered release from the nanocarriers. To confirm this, sildenafil was first encapsulated in a cisplatin-incorporated polymeric micelle. The dense PEG shell of the micelle reduced its endocytosis by cancer cells, which in return resulted in accumulative extracellular release of protonated sildenafil in the acidic tumor microenvironment. The released sildenafil was found to be more effective in enlarging the tumor blood vessels than could be achieved without sildenafil. As a result, we demonstrated considerable improvement in the intratumoral accumulation of the sildenafil-cisplatin co-loaded nanoparticle and its enhanced cancer therapeutic efficacy over the control group. Given the generality of a dense PEG shell and a hydrophobic part in most clinically developed nanomedicines, this work implies the great potential of sildenafil as a simple and universal adjuvant to selectively promote the intratumoral accumulation of nanomedicines, thus improving their clinical translation.


Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Melanoma, Experimental/metabolism , Nanoparticles/administration & dosage , Sildenafil Citrate/administration & dosage , Vasodilator Agents/administration & dosage , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Biological Transport , Blood Vessels/physiology , Cell Line, Tumor , Cisplatin/chemistry , Cisplatin/pharmacokinetics , Drug Liberation , Female , Hydrogen-Ion Concentration , Melanoma, Experimental/blood supply , Melanoma, Experimental/chemistry , Melanoma, Experimental/drug therapy , Mice, Inbred C57BL , Micelles , Nanoparticles/chemistry , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacokinetics , Polyglutamic Acid/administration & dosage , Polyglutamic Acid/analogs & derivatives , Polyglutamic Acid/chemistry , Polyglutamic Acid/pharmacokinetics , Rats, Sprague-Dawley , Sildenafil Citrate/chemistry , Sildenafil Citrate/pharmacokinetics , Tissue Distribution , Vasodilation , Vasodilator Agents/chemistry , Vasodilator Agents/pharmacokinetics
13.
PLoS One ; 15(3): e0229290, 2020.
Article En | MEDLINE | ID: mdl-32187204

Preclinical studies have shown a larger inhibition of tumour growth when exercise begins prior to tumour implant (preventative setting) than when training begins after tumour implant (therapeutic setting). However, post-implantation exercise may alter the tumour microenvironment to make it more vulnerable to treatment by increasing tumour perfusion while reducing hypoxia. This has been shown most convincingly in breast and prostate cancer models to date and it is unclear whether other tumour types respond in a similar way. We aimed to determine whether tumour perfusion and hypoxia are altered with exercise in a melanoma model, and compared this with a breast cancer model. We hypothesised that post-implantation exercise would reduce tumour hypoxia and increase perfusion in these two models. Female, 6-10 week old C57BL/6 mice were inoculated with EO771 breast or B16-F10 melanoma tumour cells before randomisation to either exercise or non-exercising control. Exercising mice received a running wheel with a revolution counter. Mice were euthanised when tumours reached maximum ethical size and the tumours assessed for perfusion, hypoxia, blood vessel density and proliferation. We saw an increase in heart to body weight ratio in exercising compared with non-exercising mice (p = 0.0008), indicating that physiological changes occurred with this form of physical activity. However, exercise did not affect vascularity, perfusion, hypoxia or tumour growth rate in either tumour type. In addition, EO771 tumours had a more aggressive phenotype than B16-F10 tumours, as inferred from a higher rate of proliferation (p<0.0001), a higher level of tumour hypoxia (p = 0.0063) and a higher number of CD31+ vessels (p = 0.0005). Our results show that although a physiological training effect was seen with exercise, it did not affect tumour hypoxia, perfusion or growth rate. We suggest that exercise monotherapy is minimally effective and that future preclinical work should focus on the combination of exercise with standard cancer therapies.


Breast Neoplasms/pathology , Melanoma, Experimental/pathology , Physical Conditioning, Animal/methods , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Melanoma, Experimental/blood supply , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Random Allocation , Running , Tumor Hypoxia , Tumor Microenvironment
14.
Sci Rep ; 10(1): 2939, 2020 02 19.
Article En | MEDLINE | ID: mdl-32076044

Anti-vascular endothelial growth factor (VEGF) therapy shows antitumor activity against various types of solid cancers. Several resistance mechanisms against anti-VEGF therapy have been elucidated; however, little is known about the mechanisms by which the acquired resistance arises. Here, we developed new anti-VEGF therapy-resistant models driven by chronic expression of the mouse VEGFR2 extracellular domain fused with the human IgG4 fragment crystallizable (Fc) region (VEGFR2-Fc). In the VEGFR2-Fc-expressing resistant tumors, we demonstrated that the FGFR2 signaling pathway was activated, and pericytes expressing high levels of FGF2 were co-localized with endothelial cells. Lenvatinib, a multiple tyrosine kinase inhibitor including VEGFR and FGFR inhibition, showed marked antitumor activity against VEGFR2-Fc-expressing resistant tumors accompanied with a decrease in the area of tumor vessels and suppression of phospho-FGFR2 in tumors. Our findings reveal the key role that intercellular FGF2 signaling between pericytes and endothelial cells plays in maintaining the tumor vasculature in anti-VEGF therapy-resistant tumors.


Angiogenesis Inhibitors/therapeutic use , Fibroblast Growth Factor 2/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Signal Transduction , Vascular Endothelial Growth Factors/antagonists & inhibitors , Angiogenesis Inhibitors/pharmacology , Animals , Cell Proliferation/drug effects , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Melanoma, Experimental/pathology , Mice , Models, Biological , Pericytes/drug effects , Pericytes/metabolism , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use , Signal Transduction/drug effects , Up-Regulation/drug effects , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factors/metabolism
15.
J Proteome Res ; 19(1): 238-247, 2020 01 03.
Article En | MEDLINE | ID: mdl-31603327

In recent years, tumor microenvironment (TME) has been recognized as potential targets for tumor treatment and the tumor vascular system is one of such targets. Fusing truncated tissue factor (tTF) with pH low insertion peptides (pHLIP), tTF-pHLIP, can target tumor vessels owing to its acidic TME and cause tumor vessel occlusion by blood clotting and subsequently effectively inhibit tumor growth. To evaluate its bioeffects, we exposed the tTF-pHLIP to normal mice and mice xenograft with B16F10 tumor and analyzed the metabolic profiling of various tissues and biofluids including plasma and urine from mice treated with and without tTF-pHLIP. A combination of nuclear magnetic resonance (NMR) and gas chromatography-mass spectrometry and ultra-high-performance liquid chromatography-mass spectrometry was employed in the study. We found that tTF-pHLIP treatment can effectively reduce tumor size and concurrently ameliorate tumor-induced alterations in the TCA cycle metabolism and lipid metabolism. In addition, we found that toxicity of tTF-pHLIP to normal mice is minor and exposure of the tTF-pHLIP induced oxidative stress to the system. Hence, we concluded that tTF-pHLIP is of low toxicity and effective in reducing tumor size as well as rebalancing tumor-induced metabolic derailment.


Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Membrane Proteins/genetics , Metabolome/drug effects , Recombinant Fusion Proteins/pharmacology , Animals , Fatty Acids/analysis , Fatty Acids/metabolism , Female , Magnetic Resonance Spectroscopy , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Recombinant Fusion Proteins/genetics , Thromboplastin/genetics , Tumor Burden , Tumor Microenvironment
16.
J Invest Dermatol ; 140(2): 445-454, 2020 02.
Article En | MEDLINE | ID: mdl-31425704

Calpains, intracellular proteases specifically inhibited by calpastatin, play a major role in neoangiogenesis involved in tumor invasiveness and metastasis. They are partly exteriorized via the ATP-binding cassette transporter A1(ABCA1) transporter, but the importance of this process in tumor growth is still unknown. The aim of our study was to investigate the role of extracellular calpains in a model of melanoma by blocking their extracellular activity or exteriorization. In the first approach, a B16-F10 model of melanoma was developed in transgenic mice expressing high extracellular levels of calpastatin. In these mice, tumor growth was inhibited by ∼ 3-fold compared with wild-type animals. In vitro cytotoxicity assays and in vivo tumor studies have demonstrated that this protection was associated with a defect in tumor neoangiogenesis. Similarly, in wild-type animals given probenecid to blunt ABCA1 activity, melanoma tumor growth was inhibited by ∼ 3-fold. Again, this response was associated with a defect in neoangiogenesis. In vitro studies confirmed that probenecid limited endothelial cell migration and capillary formation from vascular explants. The observed reduction in fibronectin cleavage under these conditions is potentially involved in the response. Collectively, these studies demonstrate that probenecid, by blunting ABCA1 activity and thereby calpain exteriorization, limits melanoma tumor neoangiogenesis and invasiveness.


ATP Binding Cassette Transporter 1/antagonists & inhibitors , Calpain/metabolism , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Probenecid/pharmacology , Skin Neoplasms/drug therapy , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line, Tumor/transplantation , Cell Proliferation/drug effects , Humans , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Mice, Transgenic , Neovascularization, Pathologic/pathology , Probenecid/therapeutic use , Skin Neoplasms/blood supply , Skin Neoplasms/pathology
17.
Int J Radiat Oncol Biol Phys ; 105(5): 1126-1136, 2019 12 01.
Article En | MEDLINE | ID: mdl-31461675

PURPOSE: Synchrotron microbeam radiation therapy (MRT) is a method that spatially distributes the x-ray beam into several microbeams of very high dose (peak dose), regularly separated by low-dose intervals (valley dose). MRT selectively spares normal tissues, relative to conventional (uniform broad beam [BB]) radiation therapy. METHODS AND MATERIALS: To evaluate the effect of MRT on radioresistant melanoma, B16-F10 murine melanomas were implanted into mice ears. Tumors were either treated with MRT (407.6 Gy peak; 6.2 Gy valley dose) or uniform BB irradiation (6.2 Gy). RESULTS: MRT induced significantly longer tumor regrowth delay than did BB irradiation. A significant 24% reduction in blood vessel perfusion was observed 5 days after MRT, and the cell proliferation index was significantly lower in melanomas treated by MRT compared with BB. MRT provoked a greater induction of senescence in melanoma cells. Bio-Plex analyses revealed enhanced concentration of monocyte-attracting chemokines in the MRT group: MCP-1 at D5, MIP-1α, MIP-1ß, IL12p40, and RANTES at D9. This was associated with leukocytic infiltration at D9 after MRT, attributed mainly to CD8 T cells, natural killer cells, and macrophages. CONCLUSIONS: In light of its potential to disrupt blood vessels that promote infiltration of the tumor by immune cells and its induction of senescence, MRT could be a new therapeutic approach for radioresistant melanoma.


Ear Neoplasms/radiotherapy , Melanoma, Experimental/radiotherapy , Radiation Tolerance , Synchrotrons , Animals , Cell Proliferation/radiation effects , Cellular Senescence , Ear Neoplasms/blood supply , Ear Neoplasms/metabolism , Ear Neoplasms/pathology , Female , Melanoma, Experimental/blood supply , Melanoma, Experimental/chemistry , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Monocyte Chemoattractant Proteins/metabolism , Staining and Labeling , Tumor Burden , Tumor Microenvironment , beta-Galactosidase
18.
Am J Pathol ; 189(10): 2102-2114, 2019 10.
Article En | MEDLINE | ID: mdl-31369756

Tumor-associated lymphangiogenesis correlates with lymph node metastasis and poor outcome in several human malignancies. In addition, the presence of functional lymphatic vessels regulates the formation of tumor inflammatory and immune microenvironments. Although lymphatic structures are often found deeply integrated into the fabric of adipose tissue, the impact of lymphangiogenesis on tumor-associated adipose tissue (AT) has not yet been investigated. Using K14-VEGFR3-Ig mice that constitutively express soluble vascular endothelial growth factor receptor (VEGFR) 3-Ig in the skin, scavenging VEGF-C and VEGF-D, the role of lymphangiogenesis in the generation of an inflammatory response within tumor-associated AT was studied. Macrophages expressing lymphatic vessel endothelial hyaluronan receptor-1 were found within peritumoral adipose tissue from melanoma-bearing K14-VEGFR3-Ig mice, which were further enriched with alternatively activated macrophages based on surface marker CD301/C-type lectin domain family 10 member A expression. The blockade of lymphangiogenesis also resulted in accumulation of the cytokine IL-6, which correlated with enhanced macrophage proliferation of the alternatively activated phenotype. Furthermore, melanomas co-implanted with freshly isolated adipose tissue macrophages grew more robustly than melanomas growing alone. In human cutaneous melanomas, adipocyte-selective FABP4 transcripts closely correlated with gene signatures of CLEC10A and were associated with poor overall survival. These data suggest that the blockade of pathways regulating lymphatic vessel formation shapes an inflammatory response within tumor-associated AT by facilitating accumulation of tumor-promoting alternatively activated macrophages.


Adipose Tissue/pathology , Inflammation/pathology , Lymphangiogenesis , Melanoma, Experimental/pathology , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Adipose Tissue/blood supply , Adipose Tissue/immunology , Animals , Female , Inflammation/immunology , Inflammation/metabolism , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Tumor Cells, Cultured , Tumor Microenvironment
19.
Mar Drugs ; 17(5)2019 Apr 29.
Article En | MEDLINE | ID: mdl-31035725

Melanoma is one of the most malignant and aggressive types of cancer worldwide. Fibroblast growth factor 2 (FGF2) is one of the critical regulators of melanoma angiogenesis and metastasis; thus, it might be an effective anti-cancer strategy to explore FGF2-targeting drug candidates from existing drugs. In this study, we evaluate the effect of the marine drug propylene glycol alginate sodium sulfate (PSS) on FGF2-mediated angiogenesis and invasion. The data shows that FGF2 selectively bound to PSS with high affinity. PSS inhibited FGF2-mediated angiogenesis in a rat aortic ring model and suppressed FGF2-mediated invasion, but not the migration of murine melanoma B16-F10 cells. The further mechanism study indicates that PSS decreased the expression of activated matrix metalloproteinase 2 (MMP-2) and matrix metalloproteinase 9 (MMP-9), and also suppressed their activity. In addition, PSS was found to decrease the level of Vimentin in B16-F10 cells, which is known to participate in the epithelial-mesenchymal transition. Notably, PSS did not elicit any changes in cancer cell viability. Based on the results above, we conclude that PSS might be a potential drug to regulate the tumor microenvironment in order to facilitate the recovery of melanoma patients.


Alginates/pharmacology , Fibroblast Growth Factor 2/metabolism , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/drug therapy , Skin Neoplasms/drug therapy , Alginates/therapeutic use , Animals , Aorta/drug effects , Aquatic Organisms/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Chick Embryo , Chorioallantoic Membrane , Drug Evaluation, Preclinical , Epithelial-Mesenchymal Transition , Humans , Laminaria/chemistry , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Neovascularization, Pathologic/pathology , Neovascularization, Physiologic/drug effects , Organ Culture Techniques , Rats , Skin Neoplasms/blood supply , Skin Neoplasms/pathology , Tumor Microenvironment/drug effects
20.
Int J Med Sci ; 16(3): 416-423, 2019.
Article En | MEDLINE | ID: mdl-30911276

Background: We recently reported that WNT10A plays a pivotal role in wound healing by regulating collagen expression/synthesis, as the depletion of WNT10A dramatically delays skin ulcer formation. WNT signaling also has a close correlation with the cancer microenvironment and proliferation, since tumors are actually considered to be 'unhealing' or 'overhealing' wounds. To ascertain the in vivo regulatory functions of WNT10A in tumor growth, we examined the net effects of WNT10A depletion using Wnt10a-deficient mice (Wnt10a -/-). Methods and Results: We subjected C57BL/6J wild-type (WT) or Wnt10a -/- mice to murine melanoma B16-F10 cell transplantation. Wnt10a -/- mice showed a significantly smaller volume of transplanted melanoma as well as fewer microvessels and less collagen expression and more necrosis than WT mice. Conclusions: Taken together, our observations suggest that critical in vivo roles of Wnt10a-depleted anti-stromagenesis prevent tumor growth, in contrast with true wound healing/scarring.


Collagen/metabolism , Melanoma, Experimental/pathology , Nerve Tissue Proteins/genetics , Skin Neoplasms/pathology , Wnt Proteins/genetics , Animals , Cell Line, Tumor , Female , Gene Deletion , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microvessels/metabolism , Microvessels/pathology , Nerve Tissue Proteins/metabolism , Skin Neoplasms/blood supply , Skin Neoplasms/metabolism , Stromal Cells/pathology , Wnt Proteins/metabolism
...