Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Mol Cancer Ther ; 22(4): 539-550, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36696581

ABSTRACT

Antibodies targeting insulin-like growth factor 1 receptor (IGF-1R) induce objective responses in only 5% to 15% of children with sarcoma. Understanding the mechanisms of resistance may identify combination therapies that optimize efficacy of IGF-1R-targeted antibodies. Sensitivity to the IGF-1R-targeting antibody TZ-1 was determined in rhabdomyosarcoma and Ewing sarcoma cell lines. Acquired resistance to TZ-1 was developed and characterized in sensitive Rh41 cells. The BRD4 inhibitor, JQ1, was evaluated as an agent to prevent acquired TZ-1 resistance in Rh41 cells. The phosphorylation status of receptor tyrosine kinases (RTK) was assessed. Sensitivity to TZ-1 in vivo was determined in Rh41 parental and TZ-1-resistant xenografts. Of 20 sarcoma cell lines, only Rh41 was sensitive to TZ-1. Cells intrinsically resistant to TZ-1 expressed multiple (>10) activated RTKs or a relatively less complex set of activated RTKs (∼5). TZ-1 decreased the phosphorylation of IGF-1R but had little effect on other phosphorylated RTKs in all resistant lines. TZ-1 rapidly induced activation of RTKs in Rh41 that was partially abrogated by knockdown of SOX18 and JQ1. Rh41/TZ-1 cells selected for acquired resistance to TZ-1 constitutively expressed multiple activated RTKs. TZ-1 treatment caused complete regressions in Rh41 xenografts and was significantly less effective against the Rh41/TZ-1 xenograft. Intrinsic resistance is a consequence of redundant signaling in pediatric sarcoma cell lines. Acquired resistance in Rh41 cells is associated with rapid induction of multiple RTKs, indicating a dynamic response to IGF-1R blockade and rapid development of resistance. The TZ-1 antibody had greater antitumor activity against Rh41 xenografts compared with other IGF-1R-targeted antibodies tested against this model.


Subject(s)
Nuclear Proteins , Sarcoma , Child , Humans , Transcription Factors , Receptor, IGF Type 1 , Sarcoma/drug therapy , Receptors, Somatomedin , Antibodies, Monoclonal/pharmacology , Cell Line, Tumor , Cell Cycle Proteins , SOXF Transcription Factors
2.
JCI Insight ; 3(16)2018 08 23.
Article in English | MEDLINE | ID: mdl-30135299

ABSTRACT

Osteosarcoma (OS), a malignant tumor of bone, kills through aggressive metastatic spread almost exclusively to the lung. Mechanisms driving this tropism for lung tissue remain unknown, though likely invoke specific interactions between tumor cells and other cells within the lung metastatic niche. Aberrant overexpression of ΔNp63 in OS cells directly drives production of IL-6 and CXCL8. All these factors were expressed at higher levels in OS lung metastases than in matched primary tumors from the same patients. Expression in cell lines correlated strongly with lung colonization efficiency in murine xenograft models. Lentivirus-mediated expression endowed poorly metastatic OS cells with increased metastatic capacity. Disruption of IL-6 and CXCL8 signaling using genetic or pharmaceutical inhibitors had minimal effects on tumor cell proliferation in vitro or in vivo, but combination treatment inhibited metastasis across multiple models of metastatic OS. Strong interactions occurred between OS cells and both primary bronchial epithelial cells and bronchial smooth muscle cells that drove feed-forward amplification of IL-6 and CXCL8 production. These results identify IL-6 and CXCL8 as primary mediators of OS lung tropism and suggest pleiotropic, redundant mechanisms by which they might effect metastasis. Combination therapy studies demonstrate proof of concept for targeting these tumor-lung interactions to affect metastatic disease.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bone Neoplasms/pathology , Interleukin-6/metabolism , Interleukin-8/metabolism , Lung Neoplasms/pathology , Adolescent , Adult , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Neoplasms/drug therapy , Bone and Bones/pathology , Cell Line, Tumor , Cell Proliferation , Child , Cytokine Receptor gp130/antagonists & inhibitors , Cytokine Receptor gp130/metabolism , Drug Evaluation, Preclinical , Follow-Up Studies , Humans , Hydrazines/pharmacology , Hydrazines/therapeutic use , Lung/pathology , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Mice , Osteosarcoma/drug therapy , Osteosarcoma/prevention & control , Osteosarcoma/secondary , Primary Cell Culture , Quinoxalines/pharmacology , Quinoxalines/therapeutic use , Receptors, Interleukin-8A/antagonists & inhibitors , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Xenograft Model Antitumor Assays , Young Adult
3.
Mol Cancer Ther ; 15(5): 1018-28, 2016 05.
Article in English | MEDLINE | ID: mdl-26908627

ABSTRACT

The bromodomain and extra-terminal domain inhibitor JQ1 has marked antitumor activity against several hematologic malignancies as well as solid tumor models. Here, we investigated its activity in vitro and in vivo against models of childhood rhabdomyosarcoma and Ewing sarcoma. In vitro, JQ1 (but not the inactive enantiomer JQ1R) inhibited cell proliferation and increased G1 fraction of cells, although there was no correlation between cell line sensitivity and suppression of c-MYC or MYCN. In vivo, xenografts showed significant inhibition of growth during the period of treatment, and rapid regrowth after treatment was stopped, activity typical of antiangiogenic agents. Furthermore, xenografts derived from cell lines intrinsically resistant or sensitive to JQ1 in vitro had similar sensitivity in vivo as xenografts. Further investigation showed that JQ1 reduced tumor vascularization. This was secondary to both drug-induced downregulation of tumor-derived growth factors and direct effects of JQ1 on vascular elements. JQ1 suppressed VEGF-stimulated vascularization of Matrigel plugs in mice, and in vitro suppressed differentiation, proliferation, and invasion of human umbilical cord vascular endothelial cells (HUVEC). In HUVECs, JQ1 partially suppressed c-MYC levels, but dramatically reduced AP-1 levels and activity through suppression of the AP-1-associated protein FOSL1. Our data suggest that the antitumor activity of JQ1 in these sarcoma models is largely a consequence of its antiangiogenic activity. Mol Cancer Ther; 15(5); 1018-28. ©2016 AACR.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Neovascularization, Pathologic/metabolism , Sarcoma/metabolism , Sarcoma/pathology , Triazoles/pharmacology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Child , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Human Umbilical Vein Endothelial Cells , Humans , Mice , Neovascularization, Pathologic/drug therapy , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Sarcoma/drug therapy , Transcription Factor AP-1/metabolism , Xenograft Model Antitumor Assays
4.
Chem Res Toxicol ; 27(12): 2015-22, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25380349

ABSTRACT

Oxidation of docosahexaenoate phospholipids produces 4-hydroxy-7-oxo-hept-5-eonyl phospholipids (HOHA-PLs) that react with protein lysyl ε-amino residues to generate 2-ω-carboxyethylpyrrole (CEP) derivatives, endogenous factors that induce angiogenesis in the retina and tumors. It seemed likely, but remained unproven, that HOHA-PLs react with ethanolamine phospholipids (EPs) in vivo to generate CEP-EPs. We now show that CEP-EPs are present in human blood at 4.6-fold higher levels in age-related macular degeneration plasma than in normal plasma. We also show that CEP-EPs are pro-angiogenic, inducing tube formation by human umbilical vein endothelial cells by activating Toll-like receptor 2. CEP-EP levels may be a useful biomarker for clinical assessment of AMD risk and CEP-associated tumor progression and a tool for monitoring the efficacy of therapeutic interventions.


Subject(s)
Phosphatidylethanolamines/blood , Phospholipids/blood , Chromatography, Liquid , Human Umbilical Vein Endothelial Cells , Humans , Macular Degeneration/blood , Magnetic Resonance Spectroscopy , Phospholipids/physiology , Tandem Mass Spectrometry
5.
Cancer Res ; 74(8): 2316-27, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24590057

ABSTRACT

STAT3 is well corroborated preclinically as a cancer therapeutic target, but tractable translational strategies for its blockade by small molecule inhibitors have remained elusive. In this study, we report the development of a novel class of bifunctional STAT3 inhibitors, based on conjugation of a diarylidenyl-piperidone (DAP) backbone to an N-hydroxypyrroline (-NOH) group, which exhibits minimal toxicity against normal cells and good oral bioavailability. Molecular modeling studies of this class suggested direct interaction with the STAT3 DNA binding domain. In particular, the DAP compound HO-3867 selectively inhibited STAT3 phosphorylation, transcription, and DNA binding without affecting the expression of other active STATs. HO-3867 exhibited minimal toxicity toward noncancerous cells and tissues but induced apoptosis in ovarian cancer cells. Pharmacologic analysis revealed greater bioabsorption and bioavailability of the active (cytotoxic) metabolites in cancer cells compared with normal cells. The selective cytotoxicity of HO-3867 seemed to be multifaceted, eliciting differential activation of the Akt pathway in normal versus cancer cells. RNAi attenuation experiments confirmed the requirement of STAT3 for HO-3867-mediated apoptosis in ovarian cancer cells. In vivo testing showed that HO-3867 could block xenograft tumor growth without toxic side effects. Furthermore, in primary human ovarian cancer cells isolated from patient ascites, HO-3867 inhibited cell migration/invasion and survival. Our results offer preclinical proof-of-concept for HO-3867 as a selective STAT3 inhibitor to treat ovarian cancer and other solid tumors where STAT3 is widely upregulated.


Subject(s)
Ovarian Neoplasms/drug therapy , Piperidones/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , CHO Cells , Cell Growth Processes/drug effects , Cell Line, Tumor , Cricetulus , Cytotoxicity, Immunologic , Female , Humans , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcriptional Activation , Transfection , Xenograft Model Antitumor Assays
6.
Oncoscience ; 1(3): 216-28, 2014.
Article in English | MEDLINE | ID: mdl-25594014

ABSTRACT

Tumor hypoxia, a feature of many solid tumors including ovarian cancer, is associated with resistance to therapies. We previously demonstrated that hypoxic exposure results in increased expression of phosphorylated signal transducer and activator of transcription 3 (pSTAT3). We hypothesized the activation of STAT3 could lead to chemotherapeutic resistance in ovarian cancer cells in hypoxic conditions. In this study, we demonstrate the level of pSTAT3 Tyr705 is increased in the hypoxic regions of human epithelial ovarian cancer (EOC) specimens, as determined by HIF-1α and CD-31 staining. In vitro mutagenesis studies proved that pSTAT3 Tyr705 is necessary for cell survival and proliferation under hypoxic conditions. In addition, we show that S1PR1, a regulator of STAT3 transcription via the JAK/STAT pathway, is highly expressed in hypoxic ovarian cancer cells (HOCCs). Knock down of S1PR1 in HOCCs reduced pSTAT3 Tyr705 levels and was associated with decreased cell survival. Treatment of HOCCs with the STAT3 inhibitor HO-3867 resulted in a rapid and dramatic decrease in pSTAT3 Tyr705 levels as a result of ubiquitin proteasome degradation. STAT3-target proteins Bcl-xL, cyclin D2 and VEGF showed similar decreases in HO-3867 treated cells. Taken together, these findings suggest that activation of STAT3 Tyr705 promotes cell survival and proliferation in HOCCs, and that S1PR1 is involved in the initiation of STAT3 activation. Targeting hypoxia-mediated STAT3 activation represents a therapeutic option for ovarian cancer and other solid tumors.

7.
Cancer Res ; 74(1): 320-9, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24154873

ABSTRACT

The tumor suppressor gene p53 and its family members p63/p73 are critical determinants of tumorigenesis. ΔNp63 is a splice variant of p63, which lacks the N-terminal transactivation domain. It is thought to antagonize p53-, p63-, and p73-dependent translation, thus blocking their tumor suppressor activity. In our studies of the pediatric solid tumors neuroblastoma and osteosarcoma, we find overexpression of ΔNp63; however, there is no correlation of ΔNp63 expression with p53 mutation status. Our data suggest that ΔNp63 itself endows cells with a gain-of-function that leads to malignant transformation, a function independent of any p53 antagonism. Here, we demonstrate that ΔNp63 overexpression, independent of p53, increases secretion of interleukin (IL)-6 and IL-8, leading to elevated phosphorylation of STAT3 (Tyr-705). We show that elevated phosphorylation of STAT3 leads to stabilization of hypoxia-inducible factor 1α (HIF-1α) protein, resulting in VEGF secretion. We also show human clinical data, which suggest a mechanistic role for ΔNp63 in osteosarcoma metastasis. In summary, our studies reveal the mechanism by which ΔNp63, as a master transcription factor, modulates tumor angiogenesis.


Subject(s)
Bone Neoplasms/blood supply , Membrane Proteins/metabolism , Neuroblastoma/blood supply , Osteosarcoma/blood supply , Adolescent , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line, Tumor , Child , Disease Models, Animal , Female , Heterografts , Humans , Membrane Proteins/genetics , Mice , Mice, SCID , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Osteosarcoma/genetics , Osteosarcoma/metabolism , Transcription Factors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
8.
J Biol Chem ; 289(7): 4083-94, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24366874

ABSTRACT

Under conditions of DNA damage, the mammalian target of rapamycin complex 1 (mTORC1) is inhibited, preventing cell cycle progression and conserving cellular energy by suppressing translation. We show that suppression of mTORC1 signaling to 4E-BP1 requires the coordinated activity of two tumor suppressors, p53 and p63. In contrast, suppression of S6K1 and ribosomal protein S6 phosphorylation by DNA damage is Akt-dependent. We find that loss of either p53, required for the induction of Sestrin 1/2, or p63, required for the induction of REDD1 and activation of the tuberous sclerosis complex, prevents the DNA damage-induced suppression of mTORC1 signaling. These data indicate that the negative regulation of cap-dependent translation by mTORC1 inhibition subsequent to DNA damage is abrogated in most human cancers.


Subject(s)
DNA Damage , Multiprotein Complexes/metabolism , Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Line, Tumor , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Neoplasms/genetics , Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , TOR Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics
9.
Clin Cancer Res ; 19(24): 6716-29, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24132923

ABSTRACT

PURPOSE: The BT-40 low-grade childhood astrocytoma xenograft model expresses mutated BRAF(V600E) and is highly sensitive to the MEK inhibitor selumetinib (AZD6244). In this study, we developed and characterized selumetinib resistance and explored approaches to circumventing the mechanisms of acquired resistance. EXPERIMENTAL DESIGN: BT-40 xenografts were selected in vivo for selumetinib resistance. Resistant tumors were obtained and characterized, as were tumors that reverted to sensitivity. Characterization included expression profiling, assessment of MEK signature and compensatory pathways, MEK inhibition, BRAF expression, and cytokine levels. Combination treatment of BT-40/AZD-resistant tumors with the MEK inhibitor and a STAT3 inhibitor (LLL12) was assessed. RESULTS: Resistance was unstable, tumors reverting to selumetinib sensitivity when passaged in untreated mice, and MEK was equally inhibited in sensitive and resistant tumors by selumetinib. Drug resistance was associated with an enhanced MEK signature and increased interleukin (IL)-6 and IL-8 expression. Selumetinib treatment induced phosphorylation of STAT3 (Y705) only in resistant xenografts, and similar results were observed in BRAF(V600E) astrocytic cell lines intrinsically resistant to selumetinib. Treatment of BT-40-resistant tumors with selumetinib or LLL12 had no significant effect, whereas combined treatment induced complete regressions of BT-40/AZD-resistant xenografts. CONCLUSIONS: Resistance to selumetinib selected in vivo in BT-40 tumor xenografts was unstable. In resistant tumors, selumetinib activated STAT3, and combined treatment with selumetinib and LLL12 induced complete responses in resistant BT-40 tumors. These results suggest dual targeting BRAF (V600E) signaling and STAT3 signaling may be effective in selumetinib-resistant tumors or may retard or prevent onset of resistance.


Subject(s)
Astrocytoma/drug therapy , Astrocytoma/genetics , Benzimidazoles/administration & dosage , MAP Kinase Kinase 1/genetics , Animals , Anthraquinones/pharmacology , Astrocytoma/pathology , Cell Line, Tumor , Child , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Interleukin-6/biosynthesis , Interleukin-8/biosynthesis , MAP Kinase Kinase 1/antagonists & inhibitors , Mice , Proto-Oncogene Proteins B-raf/genetics , STAT3 Transcription Factor/genetics , Sulfonamides/pharmacology
10.
Mol Cancer Ther ; 12(10): 1925-34, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24072884

ABSTRACT

Angiogenesis and metastasis are well recognized as processes fundamental to the development of malignancy. Both processes involve the coordination of multiple cellular and chemical activities through myriad signaling networks, providing a mass of potential targets for therapeutic intervention. This review will focus on one master regulator of cell motility, RAC1, and the existing data with regard to its role in cell motility, including particular roles for tumor angiogenesis and invasion/metastasis. We also emphasize the preclinical investigations carried out with RAC1 inhibitors to evaluate the therapeutic potential of this target. Herein, we explore potential future directions as well as the challenges of targeting RAC1 in the treatment of cancer. Recent insights at the molecular and cellular levels are paving the way for a more directed and detailed approach to target mechanisms of RAC1 regulating angiogenesis and metastasis. Understanding these mechanisms may provide insight into RAC1 signaling components as alternative therapeutic targets for tumor angiogenesis and metastasis.


Subject(s)
Molecular Targeted Therapy , Neoplasms/genetics , Neovascularization, Pathologic/genetics , rac1 GTP-Binding Protein/genetics , Cell Movement/genetics , Humans , Neoplasm Metastasis , Neoplasms/drug therapy , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Signal Transduction
11.
Clin Cancer Res ; 19(11): 2984-94, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23549869

ABSTRACT

BACKGROUND: In pediatric tumor xenograft models, tumor-derived insulin growth factor (IGF-2) results in intrinsic resistance to IGF-IR-targeted antibodies, maintaining continued tumor angiogenesis. We evaluated the antiangiogenic activity of a ligand-binding antibody (MEDI-573) alone or in combination with IGF-I receptor binding antibodies (MAB391, CP01-B02). METHODS: IGF-stimulated signaling was monitored by increased Akt phosphorylation in sarcoma and human umbilical cord vascular endothelial cells (HUVEC). Angiogenesis was determined in vitro using capillary tube formation in HUVECs and in vivo using a VEGF-stimulated Matrigel assay. Tumor growth delay was examined in 4 sarcoma xenograft models. RESULTS: The IGF ligand-binding antibody MEDI-573 suppressed Akt phosphorylation induced by exogenous IGF-I and IGF-2 in sarcoma cells. Receptor-binding antibodies suppressed IGF-I stimulation of Akt phosphorylation, but IGF-2 circumvented this effect and maintained HUVEC tube formation. MEDI-573 inhibited HUVEC proliferation and tube formation in vitro, but did not inhibit angiogenesis in vivo, probably because MEDI-573 binds murine IGF-I with low affinity. However, in vitro antiangiogenic activity of MEDI-573 was also circumvented by human recombinant IGF-I. The combination of receptor- and ligand-binding antibodies completely suppressed VEGF-stimulated proliferation of HUVECs in the presence of IGF-I and IGF-2, prevented ligand-induced phosphorylation of IGF-IR/IR receptors, and suppressed VEGF/IGF-2-driven angiogenesis in vivo. The combination of CP1-BO2 plus MEDI-573 was significantly superior to therapy with either antibody alone against IGF-I and IGF-2 secreting pediatric sarcoma xenograft models. CONCLUSIONS: These results suggest that combination of antibodies targeting IGF receptor and ligands may be an effective therapeutic strategy to block angiogenesis for IGF-driven tumors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Angiogenesis Inhibitors/administration & dosage , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Cell Proliferation/drug effects , Disease Models, Animal , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Ligands , Mice , Neovascularization, Pathologic/drug therapy , Receptor, IGF Type 1/metabolism , Sarcoma/drug therapy , Sarcoma/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
12.
PLoS One ; 7(8): e39444, 2012.
Article in English | MEDLINE | ID: mdl-22916091

ABSTRACT

The angiogenic process is controlled by variety of factors of which the vascular endothelial growth factor (VEGF) pathway plays a major role. A series of heparan sulfate mimetic small molecules targeting VEGF/VEGFR pathway has been synthesized. Among them, compound 8 (2-butyl-5-chloro-3-(4-nitro-benzyl)-3H-imidazole-4-carbaldehyde) was identified as a significant binding molecule for the heparin-binding domain of VEGF, determined by high-throughput-surface plasmon resonance assay. The data predicted strong binding of compound 8 with VEGF which may prevent the binding of VEGF to its receptor. We compared the structure of compound 8 with heparan sulfate (HS), which have in common the functional ionic groups such as sulfate, nitro and carbaldehyde that can be located in similar positions of the disaccharide structure of HS. Molecular docking studies predicted that compound 8 binds at the heparin binding domain of VEGF through strong hydrogen bonding with Lys-30 and Gln-20 amino acid residues, and consistent with the prediction, compound 8 inhibited binding of VEGF to immobilized heparin. In vitro studies showed that compound 8 inhibits the VEGF-induced proliferation migration and tube formation of mouse vascular endothelial cells, and finally the invasion of a murine osteosarcoma cell line (LM8G7) which secrets high levels of VEGF. In vivo, these effects produce significant decrease of tumor burden in an experimental model of liver metastasis. Collectively, these data indicate that compound 8 may prevent tumor growth through a direct effect on tumor cell proliferation and by inhibition of endothelial cell migration and angiogenesis mediated by VEGF. In conclusion, compound 8 may normalize the tumor vasculature and microenvironment in tumors probably by inhibiting the binding of VEGF to its receptor.


Subject(s)
Antineoplastic Agents/pharmacology , Molecular Mimicry , Vascular Endothelial Growth Factor A/metabolism , Animals , Antineoplastic Agents/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Drug Screening Assays, Antitumor , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Hydrogen Bonding , Mice , Molecular Docking Simulation , Protein Binding , Small Molecule Libraries
13.
PLoS One ; 7(4): e35513, 2012.
Article in English | MEDLINE | ID: mdl-22530037

ABSTRACT

BACKGROUND: Recent data indicate the Signal Transducer and Activator of Transcription 3 (STAT3) pathway is required for VEGF production and angiogenesis in various types of cancers. STAT3 inhibitors have been shown to reduce tumor microvessel density in tumors but a direct anti-angiogenic activity has not been described. METHODOLOGY/PRINCIPAL FINDINGS: We investigated the direct action of a small molecule inhibitor of STAT3 (LLL12) in human umbilical cord vascular endothelial cells (HUVECs) in vitro, in a Matrigel model for angiogenesis in vivo, and its antitumor activity in a xenograft model of osteosarcoma. LLL12 (100 nM) significantly inhibited VEGF-stimulated STAT3 phosphorylation in HUVECs, reduced their proliferation/migration and inhibited VEGF-induced tube formation. Morphologic analysis of LLL12 treated HUVECs demonstrated marked changes in actin/tubulin distribution and bundling. In scid mice, LLL12 reduced microvessel invasion into VEGF-infused Matrigel plugs by ∼90% at a dose of 5 mg/kg daily. Following a period of tumor progression (2 weeks), LLL12 completely suppressed further growth of established OS-1 osteosarcoma xenografts. Pharmacodynamic studies showed robust phosphorylated STAT3 in control tumors, whereas phospho-STAT3 was not detected in LLL12-treated OS-1 tumors. Treated tumors demonstrated decreased proliferation (Ki67 staining), and decreased microvessel density (CD34 staining), but no significant increase in apoptosis (TUNEL staining), relative to controls. Assay of angiogenic factors, using an antibody array, showed VEGF, MMP-9, Angiopoietin1/2, Tissue Factor and FGF-1 expression were dramatically reduced in LLL12-treated tumors compared to control tumors. CONCLUSIONS: These findings provide the first evidence that LLL12 effectively inhibits tumor angiogenesis both in vitro and in vivo.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Anthraquinones/pharmacology , Neovascularization, Physiologic/drug effects , STAT3 Transcription Factor/antagonists & inhibitors , Sulfonamides/pharmacology , Actins/metabolism , Angiogenesis Inhibitors/administration & dosage , Animals , Anthraquinones/administration & dosage , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Endothelial Cells/drug effects , Female , Humans , Mice , Mice, SCID , Microtubules/metabolism , Myocytes, Smooth Muscle/drug effects , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Phosphorylation/drug effects , Sulfonamides/administration & dosage , Vascular Endothelial Growth Factor A/pharmacology , Xenograft Model Antitumor Assays
14.
Indian J Urol ; 28(4): 377-81, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23450267

ABSTRACT

The bioactive form of vitamin D, 1α, 25-dihydroxyvitamin D3 (1α, 25(OH)2D3), is a secosteroid hormone that binds to the vitamin D receptor (VDR), a member of the nuclear receptor super-family expressed in many cell types, and modulates a variety of biological functions. 1α, 25(OH)2D3 is essential for bone and mineral homeostasis, but also regulates growth and differentiation of multiple cell types, and displays immunoregulatory and anti-inflammatory activities. The antiproliferative, prodifferentiative, antibacterial, immunomodulatory and anti-inflammatory properties of synthetic VDR agonists could be exploited to treat a variety of chronic inflammatory and autoimmune diseases, including benign prostatic hyperplasia (BPH). It has been hypothesized that VDR may influence both the risk of a variety of diseases and their occurrence and prognosis. However, earlier studies investigating the associations between specific VDR polymorphisms and various diseases often show controversial results. We performed a systematic review of the current literature on vitamin D and BPH using the PubMed and Web of Knowledge databases. The aim of this review is to summarize the current knowledge on the utility of the VDR gene regarding prostate growth as well as the pathogenesis and treatment of BPH, a complex syndrome characterized by a static component related to prostate overgrowth, a dynamic component responsible for urinary storage symptoms, and an inflammatory component. Despite the massive advances in recent decades, further research is needed to fully characterize the exact underlying mechanisms of VDR action on BPH and to comprehend how these cellular changes translate into clinical development in physical concert.

16.
Mol Cancer Ther ; 11(3): 649-59, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22188815

ABSTRACT

Previously, we reported that a predominant action of a type-1 insulin-like growth factor receptor (IGF-1R)-targeted antibody was through inhibiting tumor-derived VEGF, and indirectly, angiogenesis. Here, we examined the direct antiangiogenic activity of the IGF-1R-targeted antibody SCH717454 that inhibits ligand-receptor binding and the mechanism by which tumors circumvent its antiangiogenic activity. Inhibition of ligand-stimulated activation of IGF-1R, insulin receptor (IN-R), or downstream signaling [phosphorylation of Akt (Ser473)] was determined by receptor-specific immunoprecipitation and immunoblotting. Inhibition of angiogenesis was determined by proliferation and tube formation using human umbilical vein endothelial cells (HUVEC) in vitro and in Matrigel plugs implanted in mice. SCH717454 blocked IGF-1-stimulated but not IGF-2-stimulated phosphorylation of Akt in sarcoma cells. Immunoprecipitation using anti-IGF-1R and anti-IN-R antibodies revealed that SCH717454 equally blocked IGF-1-stimulated and IGF-2-stimulated IGF-1R phosphorylation, but not IGF-2-stimulated phosphorylation of IN-R. SCH717454 completely blocked VEGF-stimulated proliferation and tube formation of HUVECs, but exogenous IGF-2 and insulin circumvented these inhibitory effects. Coculture of HUVECs with IGF-2-secreting tumor cells completely abrogated SCH717454 inhibition of VEGF-stimulated HUVEC tube formation. In mice, SCH717454 inhibited angiogenesis in VEGF-infused Matrigel plugs, but had no inhibitory activity when plugs contained both VEGF + IGF-2. These results reveal for the first time, a role for IGF-1R signaling in VEGF-mediated angiogenesis in vitro and indicate direct antiangiogenic activity of SCH717454. Both in vitro and in vivo IGF-2 circumvented these effects through IN-R signaling. Many childhood cancers secrete IGF-2, suggesting that tumor-derived IGF-2 in the microenvironment maintains angiogenesis in the presence of IGF-1R-targeted antibodies allowing tumor progression.


Subject(s)
Antibodies, Monoclonal/pharmacology , Blood Vessels/drug effects , Insulin-Like Growth Factor II/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Angiogenesis Inhibitors/immunology , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Base Sequence , Blood Vessels/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, SCID , Molecular Sequence Data , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/prevention & control , Neovascularization, Physiologic/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor, IGF Type 1/immunology , Receptor, IGF Type 1/metabolism , Receptor, Insulin/antagonists & inhibitors , Receptor, Insulin/immunology , Receptor, Insulin/metabolism , Sarcoma/blood supply , Sarcoma/pathology , Vascular Endothelial Growth Factor A/pharmacology
17.
J Cancer Res Clin Oncol ; 137(12): 1729-38, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21932129

ABSTRACT

PURPOSE: Bladder cancer (BC) is one of the most widespread cancers afflicting men and women and also has major philosophical impact on health care worldwide. Despite elaborate characterization of the risk factors and treatment options, BC is still a major epidemiological problem worldwide and its incidence lingers to upswing each year. Over the last three decades, intravesical immunotherapy with the biological response modifier Mycobacterium bovis-Bacillus Calmette Guerin (BCG) has been established as the most effective adjuvant treatment for averting local recurrences and tumor progression following transurethral resection of non-muscle-invasive bladder cancer. DESIGN AND METHODS: PUBMED database was searched for articles, and manuscripts were selected that provided data regarding the correlation of BCG therapy and its response with different cytokine gene variants. RESULTS: It is not clear how Bacillus Calmette-Guerin (BCG) works to treat BC. It may stimulate an immune response or cause inflammation of the bladder wall that destroys cancer cells within the bladder. Lot of reports indicated the correlation of various cytokines with respect to BCG therapy in BC, but the exact mechanism is under debate. CONCLUSION: Research continues to establish the most effectual strain of BCG and the best dosage schedule for the treatment for bladder cancer but, on the other hand, a very critical part of this therapy to find out the correlation of different cytokine with BCG therapy, which will give a better insights not only the mechanism but also a better therapeutic options.


Subject(s)
BCG Vaccine/therapeutic use , Cytokines/physiology , Polymorphism, Genetic , Urinary Bladder Neoplasms/therapy , Cytokines/genetics , Female , Humans , Immunotherapy , Inflammation/complications , Male , Urinary Bladder Neoplasms/etiology , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology
18.
Sarcoma ; 2011: 601514, 2011.
Article in English | MEDLINE | ID: mdl-21197468

ABSTRACT

Angiogenesis and vasculogenesis constitute two processes in the formation of new blood vessels and are essential for progression of solid tumors. Consequently, targeting angiogenesis, and to a lesser extent vasculogenesis, has become a major focus in cancer drug development. Angiogenesis inhibitors are now being tested in pediatric populations whereas inhibitors of vasculogenesis are in an earlier stage of development. Despite the initial enthusiasm for targeting angiogenesis for treatment of cancer, clinical trials have shown only incremental increases in survival, and agents have been largely cytostatic rather than inducing tumor regressions. Consequently, the role of such therapeutic approaches in the context of curative intent for childhood sarcomas is less clear. Here we review the literature on blood vessel formation in sarcomas with a focus on pediatric sarcomas and developments in targeting angiogenesis for treatment of these rare cancers.

19.
Hypertens Res ; 33(1): 11-21, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19911001

ABSTRACT

Since their inception, angiotensin-converting enzyme (ACE) inhibitors have been used as first-line therapy for the treatment of cardiovascular and renal diseases. They restore the balance between the vasoconstrictive salt-retentive and hypertrophy-causing peptide angiotensin II (Ang II) and bradykinin, a vasodilatory and natriuretic peptide. As ACE is a promiscuous enzyme, ACE inhibitors alter the metabolism of a number of other vasoactive substances. ACE inhibitors decrease systemic vascular resistance without increasing heart rate and promote natriuresis. They have been proven effective in the treatment of hypertension, and reduce mortality in congestive heart failure and left ventricular dysfunction after myocardial infarction. They inhibit ischemic events and stabilize plaques. Furthermore, they delay the progression of diabetic nephropathy and neuropathy and act as antioxidants. Ongoing studies have elucidated protective roles for them in both memory-related disorders and cancer. Lastly, N- and C-domain selective ACE inhibitors have led to new uses for ACE inhibitors.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Cardiovascular Diseases/drug therapy , Peptidyl-Dipeptidase A/physiology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cardiovascular Diseases/physiopathology , Humans , Memory/drug effects , Renin-Angiotensin System/drug effects
20.
Eur J Med Chem ; 44(10): 3947-53, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19446930

ABSTRACT

The taxane diterpneoid 2-deacetoxytaxinine J (2-DAT-J) 1 has been isolated from the bark of Himalayan yew, Taxus baccata L. spp. wallichiana in a reasonably good yield (0.1%) and its anticancer activity against breast cancer cell lines (MCF-7 and MDA-MB-231) and normal human kidney epithelial cell line (HEK-293) has been studied. 2-DAT-J (1) showed significant in vitro activity against breast cancer cell line at a concentration of 20 microM and 10 microM in MCF-7 and MDA-MB-231 respectively. Few novel taxoids were derived (7, 8 and 10-13) from the naturally occurring 2-DAT-J (1) and screened for their anticancer activity. The structure-activity relationship studies indicated that the cinnamoyl group on C-5 and acetyl group on C-10 are essential for the anticancer activity. 2-DAT-J (1) was also tested for its in vivo activity on DMBA-induced mammary tumors in virgin female Sprague Dawley rats at a dose of 10mg/kg body weight orally for 30 days and showed significant regression in mammary tumors as compared to vehicle treated group (p<0.05).


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/drug therapy , Taxoids/chemistry , Taxoids/therapeutic use , Taxus/chemistry , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/toxicity , Breast Neoplasms/chemically induced , Cell Line , Cell Line, Tumor , Epithelial Cells/cytology , Epithelial Cells/drug effects , Female , Humans , Inhibitory Concentration 50 , Kidney/cytology , Plant Bark/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Taxoids/isolation & purification , Taxoids/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...